Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 237
Filter
1.
Cell Rep Methods ; 4(7): 100815, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38986612

ABSTRACT

The ability of cells to sense and respond to mechanical forces is critical in many physiological and pathological processes. However, determining the mechanisms by which forces affect protein function inside cells remains challenging. Motivated by in vitro demonstrations of fluorescent proteins (FPs) undergoing reversible mechanical switching of fluorescence, we investigated whether force-sensitive changes in FP function could be visualized in cells. Guided by a computational model of FP mechanical switching, we develop a formalism for its detection in Förster resonance energy transfer (FRET)-based biosensors and demonstrate its occurrence in cellulo within a synthetic actin crosslinker and the mechanical linker protein vinculin. We find that in cellulo mechanical switching is reversible and altered by manipulation of cell force generation, external stiffness, and force-sensitive bond dynamics of the biosensor. This work describes a framework for assessing FP mechanical stability and provides a means of probing force-sensitive protein function inside cells.


Subject(s)
Biosensing Techniques , Fluorescence Resonance Energy Transfer , Luminescent Proteins , Fluorescence Resonance Energy Transfer/methods , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/chemistry , Biosensing Techniques/methods , Humans , Vinculin/metabolism , Vinculin/chemistry , Actins/metabolism , Actins/chemistry , Biomechanical Phenomena
2.
Nat Commun ; 15(1): 4986, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862544

ABSTRACT

Focal adhesions form liquid-like assemblies around activated integrin receptors at the plasma membrane. How they achieve their flexible properties is not well understood. Here, we use recombinant focal adhesion proteins to reconstitute the core structural machinery in vitro. We observe liquid-liquid phase separation of the core focal adhesion proteins talin and vinculin for a spectrum of conditions and interaction partners. Intriguingly, we show that binding to PI(4,5)P2-containing membranes triggers phase separation of these proteins on the membrane surface, which in turn induces the enrichment of integrin in the clusters. We suggest a mechanism by which 2-dimensional biomolecular condensates assemble on membranes from soluble proteins in the cytoplasm: lipid-binding triggers protein activation and thus, liquid-liquid phase separation of these membrane-bound proteins. This could explain how early focal adhesions maintain a structured and force-resistant organization into the cytoplasm, while still being highly dynamic and able to quickly assemble and disassemble.


Subject(s)
Cell Membrane , Focal Adhesions , Talin , Vinculin , Talin/metabolism , Talin/chemistry , Focal Adhesions/metabolism , Cell Membrane/metabolism , Vinculin/metabolism , Vinculin/chemistry , Humans , Animals , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Integrins/metabolism , Integrins/chemistry , Cytoplasm/metabolism , Protein Binding , Phase Separation
3.
J Biomed Opt ; 28(8): 082808, 2023 08.
Article in English | MEDLINE | ID: mdl-37441563

ABSTRACT

Significance: Forces inside cells play a fundamental role in tissue growth, affecting important processes such as cancer cell migration or tissue repair after injury. Förster resonance energy transfer (FRET)-based tension sensors are a remarkable tool for studying these forces and should be made easier to use. Aim: We prove that absolute FRET efficiency can be measured on a simple setup, an order of magnitude more cost-effective than a standard FRET microscopy setup, by applying it to vinculin tension sensors (VinTS) at the focal adhesions of live CHO-K1 cells. Approach: Our setup located at Université Paris-Saclay acquires donor and acceptor fluorescence in parallel on two low-cost CMOS cameras and uses two LEDs for rapid switching of the excitation wavelength at a reduced cost. The calibration required to extract FRET efficiency was achieved using a single construct (TSMod). FRET efficiencies were measured for VinTS and the tail-less control VinTL, lacking the actin-binding domain of vinculin. Measurements were confirmed on the same cell type using a more standard intensity-based setup located at Rutgers University. Results: The average FRET efficiency of VinTS (22.0%±4%) over more than 10,000 focal adhesions is significantly lower (p<10-6) than that of VinTL (30.4%±5%), our control that is insensitive to force, in agreement with the force exerted on vinculin at focal adhesions. Attachment of the CHO-K1 cells on fibronectin decreases FRET efficiency, thus increasing the force, compared with poly-lysine. FRET efficiency for the VinTL control is consistent with all measurements currently available in the literature, confirming the validity of our measurements and hence of our simpler setup. Conclusions: Force measurements, resolved spatially inside a cell, can be achieved using FRET-based tension sensors with a cost effective intensity-based setup. This will facilitate combining FRET with techniques for applying controlled forces such as optical tweezers.


Subject(s)
Fluorescence Resonance Energy Transfer , Focal Adhesions , Humans , Fluorescence Resonance Energy Transfer/methods , Focal Adhesions/metabolism , Vinculin/chemistry , Cost-Benefit Analysis , Mechanical Phenomena
4.
J Mol Recognit ; 36(6): e3012, 2023 06.
Article in English | MEDLINE | ID: mdl-36987702

ABSTRACT

Vinculin is an integral component of integrin adhesions, where it functions as a molecular clutch coupling intracellular contraction to the extracellular matrix. Quantitating its contribution to the reinforcement of newly forming adhesions, however, requires ultrasensitive cell force assays covering short time and low force ranges. Here, we have combined atomic force microscopy-based single-cell force spectroscopy (SCFS) and optical tweezers force spectroscopy to investigate the role of vinculin in reinforcement of individual nascent adhesions during the first 5 min of cell contact with fibronectin or vitronectin. At minimal adhesion times (5-10 s), mouse embryonic fibroblast (MEF) wildtype (wt) and vinculin knock-out (vin(-/-) ) cells develop comparable adhesion forces on the scale of several individual integrin-ligand bonds, confirming that vinculin is dispensable for adhesion initiation. In contrast, after 60 to 120 s, adhesion strength and traction reinforce quickly in wt cells, while remaining low in vin(-/-) cells. Re-expression of full-length vinculin or a constitutively active vinculin mutant (vinT12) in MEF vin(-/-) cells restored adhesion and traction with the same efficiency, while vinculin with a mutated talin-binding head region (vinA50I) or missing the actin-binding tail-domain (vin880) was ineffective. Integrating total internal reflection fluorescence imaging into the SCFS setup furthermore enabled us to correlate vinculin-green fluorescent protein (GFP) recruitment to nascent adhesion sites with the built-up of vinculin-dependent adhesion forces directly. Vinculin recruitment and cell adhesion reinforcement followed synchronous biphasic patterns, suggesting vinculin recruitment, but not activation, as the rate-limiting step for adhesion reinforcement. Combining sensitive SCFS with fluorescence microscopy thus provides insight into the temporal sequence of vinculin-dependent mechanical reinforcement in nascent integrin adhesions.


Subject(s)
Fibroblasts , Focal Adhesions , Animals , Mice , Cell Adhesion/physiology , Fibroblasts/metabolism , Focal Adhesions/metabolism , Integrins/metabolism , Talin/genetics , Talin/chemistry , Talin/metabolism , Vinculin/genetics , Vinculin/chemistry , Vinculin/metabolism
5.
J Mol Biol ; 435(5): 167969, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36682678

ABSTRACT

Classical cadherins are transmembrane proteins whose extracellular domains link neighboring cells, and whose intracellular domains connect to the actin cytoskeleton via ß-catenin and α-catenin. The cadherin-catenin complex transmits forces that drive tissue morphogenesis and wound healing. In addition, tension-dependent changes in αE-catenin conformation enables it to recruit the actin-binding protein vinculin to cell-cell junctions, which contributes to junctional strengthening. How and whether multiple cadherin-complexes cooperate to reinforce cell-cell junctions in response to load remains poorly understood. Here, we used single-molecule optical trap measurements to examine how multiple cadherin-catenin complexes interact with F-actin under load, and how this interaction is influenced by the presence of vinculin. We show that force oriented toward the (-) end of the actin filament results in mean lifetimes 3-fold longer than when force was applied towards the barbed (+) end. We also measured force-dependent actin binding by a quaternary complex comprising the cadherin-catenin complex and the vinculin head region, which cannot itself bind actin. Binding lifetimes of this quaternary complex increased as additional complexes bound F-actin, but only when load was oriented toward the (-) end. In contrast, the cadherin-catenin complex alone did not show this form of cooperativity. These findings reveal multi-level, force-dependent regulation that enhances the strength of the association of multiple cadherin/catenin complexes with F-actin, conferring positive feedback that may strengthen the junction and polarize F-actin to facilitate the emergence of higher-order cytoskeletal organization.


Subject(s)
Actin Cytoskeleton , Actins , Vinculin , alpha Catenin , Actin Cytoskeleton/metabolism , Actins/metabolism , alpha Catenin/chemistry , alpha Catenin/metabolism , Cadherins/chemistry , Cadherins/metabolism , Cell Adhesion , Protein Binding , Vinculin/chemistry , Allosteric Regulation
6.
Protein J ; 41(1): 55-70, 2022 02.
Article in English | MEDLINE | ID: mdl-35006498

ABSTRACT

To shed light onto the activation mechanism of vinculin, we carried out a detailed refinement of chicken vinculin and compared it to the human protein which is greater than 95% identical. Refinement resulted in a complete and significantly improved model. This model includes important elements such as a pro-rich strap region (PRR) and C-terminus. The conformation of the PRR stabilized by its inter- and intra-molecular contacts shows a dynamic, but relatively stable motif that constitutes a docking platform for multiple molecules. The contact of the C-terminus with the PRR suggests that phosphorylation of Tyr1065 might control activation and membrane binding. Improved electron densities showed the presence of large solvent molecules such as phosphates/sulfates and a head-group of PIP2. The improved model allowed for a computational stability analysis to be performed by the program Corex/Best which located numerous hot-spots of increased and decreased stability. Proximity of the identified binding sites for regulatory partners involved in inducing or suppressing the activation of vinculin to the unstable elements sheds new light onto the activation pathway and differential activation. This stability analysis suggests that the activation pathway proceeds by unfurling of the super-bundle built from four bundles of helices without separation of the Vt region (840-1066) from the head. According to our mechanism, when activating proteins bind at the strap region a separation of N and C terminal bundles occurs, followed by unfurling of the super-bundle and flattening of the general shape of the molecule, which exposes the interaction sites for binding of auxiliary proteins.


Subject(s)
Actins , Vinculin , Actins/chemistry , Animals , Binding Sites , Chickens , Humans , Protein Binding , Protein Conformation , Vinculin/chemistry
7.
PLoS One ; 16(5): e0251411, 2021.
Article in English | MEDLINE | ID: mdl-33974655

ABSTRACT

Cells exert traction forces on the extracellular matrix to which they are adhered through the formation of focal adhesions. Spatial-temporal regulation of traction forces is crucial in cell adhesion, migration, cellular division, and remodeling of the extracellular matrix. By cultivating cells on polyacrylamide hydrogels of different stiffness we were able to investigate the effects of substrate stiffness on the generation of cellular traction forces by Traction Force Microscopy (TFM), and characterize the molecular dynamics of the focal adhesion protein zyxin by Fluorescence Correlation Spectroscopy (FCS) and Fluorescence Recovery After Photobleaching (FRAP). As the rigidity of the substrate increases, we observed an increment of both, cellular traction generation and zyxin residence time at the focal adhesions, while its diffusion would not be altered. Moreover, we found a positive correlation between the traction forces exerted by cells and the residence time of zyxin at the substrate elasticities studied. We found that this correlation persists at the subcellular level, even if there is no variation in substrate stiffness, revealing that focal adhesions that exert greater traction present longer residence time for zyxin, i.e., zyxin protein has less probability to dissociate from the focal adhesion.


Subject(s)
Stress, Mechanical , Zyxin/chemistry , Actin Cytoskeleton/drug effects , Amides/pharmacology , Animals , Cattle , Cell Adhesion , Cytochalasin D/pharmacology , Endothelial Cells , Fluorescence Recovery After Photobleaching , Focal Adhesions , Green Fluorescent Proteins , Intravital Microscopy , Kinetics , Lasers , Mice , Mice, Inbred BALB C , Pyridines/pharmacology , Recombinant Fusion Proteins/chemistry , Vinculin/chemistry , rho-Associated Kinases/antagonists & inhibitors
8.
Int J Mol Sci ; 22(2)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440717

ABSTRACT

Vinculin and its heart-specific splice variant metavinculin are key regulators of cell adhesion processes. These membrane-bound cytoskeletal proteins regulate the cell shape by binding to several other proteins at cell-cell and cell-matrix junctions. Vinculin and metavinculin link integrin adhesion molecules to the filamentous actin network. Loss of both proteins prevents cell adhesion and cell spreading and reduces the formation of stress fibers, focal adhesions, or lamellipodia extensions. The binding of talin at cell-matrix junctions or of α-catenin at cell-cell junctions activates vinculin and metavinculin by releasing their autoinhibitory head-tail interaction. Once activated, vinculin and metavinculin bind F-actin via their five-helix bundle tail domains. Unlike vinculin, metavinculin has a 68-amino-acid insertion before the second α-helix of this five-helix F-actin-binding domain. Here, we present the full-length cryogenic electron microscopy structure of metavinculin that captures the dynamics of its individual domains and unveiled a hallmark structural feature, namely a kinked isoform-specific α-helix in its F-actin-binding domain. Our identified conformational landscape of metavinculin suggests a structural priming mechanism that is consistent with the cell adhesion functions of metavinculin in response to mechanical and cellular cues. Our findings expand our understanding of metavinculin function in the heart with implications for the etiologies of cardiomyopathies.


Subject(s)
Cryoelectron Microscopy , Cytoskeleton/chemistry , Protein Interaction Domains and Motifs , Vinculin/chemistry , Actins/chemistry , Actins/metabolism , Cell Adhesion , Cell Membrane/chemistry , Cell Membrane/metabolism , Crystallography, X-Ray , Cytoskeleton/metabolism , Humans , Models, Molecular , Peptides , Protein Binding , Protein Conformation , Structure-Activity Relationship , Vinculin/metabolism
9.
J Biomater Appl ; 35(6): 579-591, 2021 01.
Article in English | MEDLINE | ID: mdl-32847463

ABSTRACT

Keratin-based biomaterials represent an attractive opportunity in the fields of wound healing and tissue regeneration, not only for their chemical and physical properties, but also for their ability to act as a delivery system for a variety of payloads. Importantly, keratins are the only natural biomaterial that is not targeted by specific tissue turnover-related enzymes, giving it potential stability advantages and greater control over degradation after implantation. However, in-situ polymerization chemistry in some keratin systems are not compatible with cells, and incorporation within constructs such as hydrogels may lead to hypoxia and cell death. To address these challenges, we envisioned a pre-formed keratin microparticle on which cells could be seeded, while other payloads (e.g. drugs, growth factors or other biologic compounds) could be contained within, although studies investigating the potential partitioning between phases during emulsion polymerization would need to be conducted. This study employs well-established water-in-oil emulsion procedures as well as a suspension culture method to load keratin-based microparticles with bone marrow-derived mesenchymal stem cells. Fabricated microparticles were characterized for size, porosity and surface structure and further analyzed to investigate their ability to form gels upon hydration. The suspension culture technique was validated based on the ability for loaded cells to maintain their viability and express actin and vinculin proteins, which are key indicators of cell attachment and growth. Maintenance of expression of markers associated with cell plasticity was also investigated. As a comparative model, a collagen-coated microparticle (Sigma) of similar size was used. Results showed that an oxidized form of keratin ("keratose" or "KOS") formed unique microparticle structures of various size that appeared to contain a fibrous sub-structure. Cell adhesion and viability was greater on keratin microparticles compared to collagen-coated microparticles, while marker expression was retained on both.


Subject(s)
Coated Materials, Biocompatible/chemistry , Hydrogels/chemistry , Keratins/chemistry , Tissue Scaffolds/chemistry , Actins/chemistry , Cell Adhesion , Collagen/chemistry , Humans , Keratins/pharmacology , Mesenchymal Stem Cells/metabolism , Microspheres , Porosity , Surface Properties , Tissue Engineering , Vinculin/chemistry , Wound Healing/drug effects
10.
Lab Invest ; 100(8): 1030-1041, 2020 08.
Article in English | MEDLINE | ID: mdl-32238906

ABSTRACT

Talin and vinculin, both actin-cytoskeleton-related proteins, have been documented to participate in establishing bacterial infections, respectively, as the adapter protein to mediate cytoskeleton-driven dynamics of the plasma membrane. However, little is known regarding the potential role of the talin-vinculin complex during spotted fever group rickettsial and Ebola virus infections, two dreadful infectious diseases in humans. Many functional properties of proteins are determined by their participation in protein-protein complexes, in a temporal and/or spatial manner. To resolve the limitation of application in using mouse primary antibodies on archival, multiple formalin-fixed mouse tissue samples, which were collected from experiments requiring high biocontainment, we developed a practical strategic proximity ligation assay (PLA) capable of employing one primary antibody raised in mouse to probe talin-vinculin spatial proximal complex in mouse tissue. We observed an increase of talin-vinculin spatial proximities in the livers of spotted fever Rickettsia australis or Ebola virus-infected mice when compared with mock mice. Furthermore, using EPAC1-knockout mice, we found that deletion of EPAC1 could suppress the formation of spatial proximal complex of talin-vinculin in rickettsial infections. In addition, we observed increased colocalization between spatial proximity of talin-vinculin and filamentous actin-specific phalloidin staining in single survival mouse from an ordinarily lethal dose of rickettsial or Ebola virus infection. These findings may help to delineate a fresh insight into the mechanisms underlying liver specific pathogenesis during infection with spotted fever rickettsia or Ebola virus in the mouse model.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Hemorrhagic Fever, Ebola/metabolism , Liver/metabolism , Talin/metabolism , Vinculin/metabolism , Animals , Cells, Cultured , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Liver/microbiology , Liver/virology , Mice, Knockout , Protein Binding , Rickettsia/physiology , Spotted Fever Group Rickettsiosis/metabolism , Spotted Fever Group Rickettsiosis/microbiology , Talin/chemistry , Vinculin/chemistry
11.
Biochim Biophys Acta Gen Subj ; 1864(2): 129419, 2020 02.
Article in English | MEDLINE | ID: mdl-31449838

ABSTRACT

Structural study of multidomain proteins using NMR is an emerging issue for understanding biological functions. To this end, domain-specific labeling is expected to be a key technology for facilitating the NMR-assignment process and for collecting distance information via spin labeling. To obtain domain-specific labeled samples, use of sortase A as a protein ligation tool is a viable approach. Sortase A enables ligation of separately expressed proteins (domains) through the Leu-Pro-X-Thr-Gly linker. However, the ligation reaction mediated by sortase A is not efficient. Poor yield and long reaction times hamper large-scale preparation using sortase A. Here we report the application of highly active sortases to NMR analyses. Optimal yields can be achieved within several hours when the ligation reaction are mediated by highly active sortases at 4 °C. We propose that this protocol can contribute to structural analyses of multidomain proteins by NMR.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Cysteine Endopeptidases/chemistry , Staphylococcus aureus/enzymology , Escherichia coli , Hydrolysis , Magnetic Resonance Spectroscopy , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Protein Binding , Protein Domains , Protein Interaction Mapping , Temperature , Vinculin/chemistry
12.
Biochim Biophys Acta Gen Subj ; 1864(1): 129450, 2020 01.
Article in English | MEDLINE | ID: mdl-31676296

ABSTRACT

BACKGROUND: Leucine rich Aspartate motifs (LD motifs) are molecular recognition motifs on Paxillin that recognize LD-motif binding domains (LDBD) of a number of focal adhesion proteins in order to carry out downstream signaling and actin cytoskeleton remodeling. In this study, we identified structural features within LDBDs that influence their binding affinity with Paxillin LD motifs. METHODS: Various point mutants of focal adhesion targeting (FAT) domain of Focal Adhesion Kinase (FAK) were created by moving a key Lysine residue two and three helical turns in order to match the unique conformations as observed in LDBDs of two other focal adhesion proteins, Vinculin and CCM3. RESULTS: This led to identify a mutant of FAT domain of FAK, named as FAT(NV) (Asn992 of FAT domain was replaced by Val), with remarkable high affinity for LD1 (Kd = 1.5 µM vs no-binding with wild type) and LD2 peptides (Kd = 7.2 µM vs 63 µM with wild type). Consistently, the focal adhesions of MCF7 cells expressing FAK(NV) were highly stable (turnover rate = 1.25 × 10-5 µm2/s) as compared to wild type FAK transfected cells (turnover rate = 1.5 × 10-3 µm2/s). CONCLUSIONS: We observed that the relative disposition of key LD binding amino-acids at LDBD surface, hydrophobic burial of long Leucine side chains of LD-motifs and complementarity of charged surfaces are the key factors determining the binding affinities of LD motifs with LDBDs. GENERAL SIGNIFICANCE: Our study will help in protein engineering of FAT domain of FAK by modulating FAK-LD motif interactions which have implications in cellular focal adhesions and cell migration.


Subject(s)
Cell Adhesion/genetics , Focal Adhesion Kinase 1/genetics , Focal Adhesions/genetics , Protein Conformation , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Amino Acid Motifs/genetics , Amino Acid Sequence/genetics , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Aspartic Acid/genetics , Binding Sites/genetics , Cell Movement/genetics , Focal Adhesion Kinase 1/chemistry , Focal Adhesions/chemistry , Gene Expression Regulation/genetics , Humans , Lysine/chemistry , Lysine/genetics , MCF-7 Cells , Membrane Proteins/chemistry , Membrane Proteins/genetics , Paxillin/chemistry , Paxillin/genetics , Protein Binding/genetics , Protein Engineering , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Vinculin/chemistry , Vinculin/genetics
13.
J Biomed Opt ; 24(12): 1-11, 2019 12.
Article in English | MEDLINE | ID: mdl-31884745

ABSTRACT

We demonstrate the possibility of measuring FRET efficiency with a low-cost frequency-domain fluorescence lifetime imaging microscope (FD-FLIM). The system utilizes single-frequency-modulated excitation, which enables the use of cost-effective laser sources and electronics, simplification of data acquisition and analysis, and a dual-channel detection capability. Following calibration with coumarin 6, we measured the apparent donor lifetime in mTFP1-mVenus FRET standards expressed in living cells. We evaluated the system's sensitivity by differentiating the short and long lifetimes of mTFP1 corresponding to the known standards' high and low FRET efficiency, respectively. Furthermore, we show that the lifetime of the vinculin tension sensor, VinTS, at focal adhesions (2.30 ± 0.16 ns) is significantly (p < 10 - 6) longer than the lifetime of the unloaded TSMod probe (2.02 ± 0.16 ns). The pixel dwell time was 6.8 µs for samples expressing the FRET standards, with signal typically an order of magnitude higher than VinTS. The apparent FRET efficiency (EFRETapp) of the standards, calculated from the measured apparent lifetime, was linearly related to their known FRET efficiency by a factor of 0.92 to 0.99 (R2 = 0.98). This relationship serves as a calibration curve to convert apparent FRET to true FRET and circumvent the need to measure multiexponential lifetime decays. This approach yielded a FRET efficiency of 18% to 19.5%, for VinTS, in agreement with published values. Taken together, our results demonstrate a cost-effective, fast, and sensitive FD-FLIM approach with the potential to facilitate applications of FLIM in mechanobiology and FRET-based biosensing.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Microscopy, Fluorescence/methods , Molecular Probes/chemistry , Animals , Cell Line , Equipment Design , Fluorescence Resonance Energy Transfer/instrumentation , Focal Adhesions/physiology , Image Processing, Computer-Assisted , Mice , Microscopy, Fluorescence/instrumentation , Signal Processing, Computer-Assisted , Vinculin/chemistry
14.
PLoS One ; 14(9): e0221962, 2019.
Article in English | MEDLINE | ID: mdl-31483833

ABSTRACT

Vinculin (Vcn) is a ubiquitously expressed cytoskeletal protein that links transmembrane receptors to actin filaments, and plays a key role in regulating cell adhesion, motility, and force transmission. Metavinculin (MVcn) is a Vcn splice isoform that contains an additional exon encoding a 68-residue insert within the actin binding tail domain. MVcn is selectively expressed at sub-stoichiometic amounts relative to Vcn in smooth and cardiac muscle cells. Mutations in the MVcn insert are linked to various cardiomyopathies. In vitro analysis has previously shown that while both proteins can engage filamentous (F)-actin, only Vcn can promote F-actin bundling. Moreover, we and others have shown that MVcn can negatively regulate Vcn-mediated F-actin bundling in vitro. To investigate functional differences between MVcn and Vcn, we stably expressed either Vcn or MVcn in Vcn-null mouse embryonic fibroblasts. While both MVcn and Vcn were observed at FAs, MVcn-expressing cells had larger but fewer focal adhesions per cell compared to Vcn-expressing cells. MVcn-expressing cells migrated faster and exhibited greater persistence compared to Vcn-expressing cells, even though Vcn-containing FAs assembled and disassembled faster. Magnetic tweezer measurements on Vcn-expressing cells show a typical cell stiffening phenotype in response to externally applied force; however, this was absent in Vcn-null and MVcn-expressing cells. Our findings that MVcn expression leads to larger but fewer FAs per cell, in conjunction with the inability of MVcn to bundle F-actin in vitro and rescue the cell stiffening response, are consistent with our previous findings of actin bundling deficient Vcn variants, suggesting that deficient actin-bundling may account for some of the differences between Vcn and MVcn.


Subject(s)
Cell Movement , Focal Adhesions , Mechanotransduction, Cellular , Vinculin/metabolism , Animals , Cell Line , Gene Expression Regulation , Mice , Models, Molecular , Protein Domains , Vinculin/chemistry
15.
Structure ; 27(10): 1527-1536.e3, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31422909

ABSTRACT

Vinculin and its splice isoform metavinculin play key roles in regulating cellular morphology, motility, and force transduction. Vinculin is distinct from metavinculin in its ability to bundle filamentous actin (F-actin). To elucidate the molecular basis for these differences, we employed computational and experimental approaches. Results from these analyses suggest that the C terminus of both vinculin and metavinculin form stable interactions with the F-actin surface. However, the metavinculin tail (MVt) domain contains a 68 amino acid insert, with helix 1 (H1) sequestered into a globular subdomain, which protrudes from the F-actin surface and prevents actin bundling by sterically occluding actin filaments. Consistent with our model, deletion and selective point mutations within the MVt H1 disrupt this protruding structure, and facilitate actin bundling similar to vinculin tail (Vt) domain.


Subject(s)
Vinculin/chemistry , Vinculin/metabolism , Actins , Alternative Splicing , Animals , Binding Sites , Cryoelectron Microscopy , Models, Molecular , Mutation , Protein Binding , Protein Domains , Protein Structure, Secondary , Vinculin/genetics
16.
J Mol Biol ; 431(8): 1604-1618, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30844403

ABSTRACT

Debilitating heart conditions, notably dilated and hypertrophic cardiomyopathies (CMs), are associated with point mutations in metavinculin, a larger isoform of the essential cytoskeletal protein vinculin. Metavinculin is co-expressed with vinculin at sub-stoichiometric ratios in cardiac tissues. CM mutations in the metavinculin tail domain (MVt) occur within the extra 68-residue insert that differentiates it from the vinculin tail domain (Vt). Vt binds actin filaments (F-actin) and promotes vinculin dimerization to bundle F-actin into thick fibers. While MVt binds to F-actin in a similar manner to Vt, MVt is incapable of F-actin bundling and inhibits Vt-mediated F-actin bundling. We performed F-actin co-sedimentation and negative-stain EM experiments to dissect the coordinated roles of metavinculin and vinculin in actin fiber assembly and the effects of three known metavinculin CM mutations. These CM mutants were found to weakly induce the formation of disordered F-actin assemblies. Notably, they fail to inhibit Vt-mediated F-actin bundling and instead promote formation of large assemblies embedded with linear bundles. Computational models of MVt bound to F-actin suggest that MVt undergoes a conformational change licensing the formation of a protruding sub-domain incorporating the insert, which sterically prevents dimerization and bundling of F-actin by Vt. Sub-domain formation is destabilized by CM mutations, disrupting this inhibitory mechanism. These findings provide new mechanistic insights into the ability of metavinculin to tune actin organization by vinculin and suggest that dysregulation of this process by CM mutants could underlie their malfunction in disease.


Subject(s)
Actins/metabolism , Cardiomyopathies/genetics , Point Mutation , Vinculin/genetics , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Chickens , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Interaction Maps , Vinculin/chemistry , Vinculin/metabolism
17.
Sci Adv ; 5(12): eaav2720, 2019 12.
Article in English | MEDLINE | ID: mdl-31897422

ABSTRACT

The vinculin-mediated mechanosensing requires establishment of stable mechanical linkages between vinculin to integrin at focal adhesions and to cadherins at adherens junctions through associations with the respective adaptor proteins talin and α-catenin. However, the mechanical stability of these critical vinculin linkages has yet to be determined. Here, we developed a single-molecule detector assay to provide direct quantification of the mechanical lifetime of vinculin association with the vinculin binding sites in both talin and α-catenin, which reveals a surprisingly high mechanical stability of the vinculin-talin and vinculin-α-catenin interfaces that have a lifetime of >1000 s at forces up to 10 pN and can last for seconds to tens of seconds at 15 to 25 pN. Our results suggest that these force-bearing intermolecular interfaces provide sufficient mechanical stability to support the vinculin-mediated mechanotransduction at cell-matrix and cell-cell adhesions.


Subject(s)
Mechanotransduction, Cellular/physiology , Single Molecule Imaging/methods , Talin/metabolism , Vinculin/metabolism , alpha Catenin/metabolism , Actin Cytoskeleton/metabolism , Binding Sites , Cell Adhesion/physiology , Focal Adhesions/metabolism , Humans , Integrins/metabolism , Plasmids/genetics , Protein Binding , Vinculin/chemistry
18.
J Cell Sci ; 132(2)2019 01 21.
Article in English | MEDLINE | ID: mdl-30578314

ABSTRACT

Extracellular matrix (ECM) stiffness regulates various cell behaviors, including cell differentiation, proliferation and migration. Vinculin and vinexin α (an isoform encoded by the SORBS3 gene), both of which localize to focal adhesions, cooperatively function as mechanosensors of ECM stiffness. On a rigid ECM, vinexin α interacts with vinculin and induces a conformational change in vinculin to give an 'open' form, which promotes nuclear localization of Yes-associated protein (YAP, also known as YAP1) and transcriptional coactivator with a PDZ-binding motif (TAZ, also known as WWTR1) (hereafter YAP/TAZ). However, the detailed mechanism by which vinexin α induces the conformational change in vinculin has not been revealed. Here, we identify an amphipathic helix named H2 as a novel vinculin-binding site in vinexin α. The H2 helix interacts with the vinculin D1b subdomain and promotes the formation of a talin-vinculin-vinexin α ternary complex. Mutations in the H2 region not only impair the ability of vinexin α to induce the ECM stiffness-dependent conformational change in vinculin but also to promote nuclear localization of YAP/TAZ on rigid ECM. Taken together, these results demonstrate that the H2 helix in vinexin α plays a critical role in ECM stiffness-dependent regulation of vinculin and cell behaviors.


Subject(s)
Extracellular Matrix/metabolism , Muscle Proteins/metabolism , Vinculin/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Mice , Muscle Proteins/chemistry , Muscle Proteins/genetics , Protein Structure, Secondary , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism , Vinculin/chemistry , Vinculin/genetics , YAP-Signaling Proteins
19.
ACS Appl Mater Interfaces ; 10(43): 36776-36785, 2018 Oct 31.
Article in English | MEDLINE | ID: mdl-30284813

ABSTRACT

The biomimetic anisotropic particles have different physicochemical properties on the opposite two sides, enabling diverse applications in emulsion, photonic display, and diagnosis. However, the traditional anisotropic particles have a very small size, ranging from submicrons to a few microns. The design and fabrication of anisotropic macron-sized particles with new structures and properties is still challenging. In this study, anisotropic polycaprolactone (PCL) microparticles well separated with each other were prepared by crystallization from the dilute PCL solution in a porous 3D gelatin template. They had fuzzy and smooth surfaces on each side, and a size as large as 70 µm. The fuzzy surface of the particle adsorbed significantly larger amount of proteins, and was more cell-attractive regardless of the cell types. The particles showed stronger affinity toward fibroblasts over hepatocytes, which paved a new way for cell isolation merely based on the surface morphology. After a successive seeding process, Janus cell microparticles with fibroblasts and endothelial cells (ECs) on each side were designed and obtained by making use of the anisotropic surface morphology, which showed significant difference in EC functions in terms of prostacyclin (PGl2) secretion, demonstrating the unique and appealing functions of this type of anisotropic microspheres.


Subject(s)
Anisotropy , Biocompatible Materials/chemistry , Biomimetic Materials , Cell Adhesion , Microspheres , Adsorption , Animals , Cattle , Cyclin D1/chemistry , Hepatocytes/metabolism , Integrin beta1/chemistry , Materials Testing , Mice , NIH 3T3 Cells , Particle Size , Photons , Polyesters/chemistry , Serum Albumin/chemistry , Surface Properties , Vinculin/chemistry
20.
FEBS Lett ; 592(10): 1751-1760, 2018 05.
Article in English | MEDLINE | ID: mdl-29710402

ABSTRACT

Vinculin is a central component of mechanosensitive adhesive complexes that form between cells and the extracellular matrix. A myriad of infectious agents mimic vinculin binding sites (VBS), enabling them to hijack the adhesion machinery and facilitate cellular entry. Here, we report the structural and biochemical characterisation of VBS from the chlamydial virulence factor TarP. Whilst the affinities of isolated VBS peptides from TarP and talin for vinculin are similar, their behaviour in larger fragments is markedly different. In talin, VBS are cryptic and require mechanical activation to bind vinculin, whereas the TarP VBS are located in disordered regions, and so are constitutively active. We demonstrate that the TarP VBS can uncouple talin:vinculin complexes, which may lead to adhesion destabilisation.


Subject(s)
Bacterial Proteins/physiology , Chlamydia/pathogenicity , Molecular Mimicry , Talin/physiology , Vinculin/metabolism , Virulence Factors/physiology , Amino Acid Sequence , Crystallography, X-Ray , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Talin/chemistry , Talin/metabolism , Vinculin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL