Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
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
2.
STAR Protoc ; 2(2): 100449, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33997802

ABSTRACT

This protocol for the separation of nuclear and cytoplasmic fractions of cells of Xenopus laevis embryos was developed to study changes in the intracellular localization of the Zyxin and Ybx1 proteins, which are capable of changing localization in response to certain stimuli. Western blot analysis allows the quantification of changes in the distribution of these proteins between the cytoplasm and nucleus, whereas the posttranslational modifications specific to each compartment can be identified by changes in electrophoretic mobility. For complete details on the use and execution of this protocol, please refer to Parshina et al. (2020).


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Embryo, Nonmammalian/cytology , Xenopus Proteins , Xenopus laevis/embryology , Animals , Female , Male , Xenopus Proteins/analysis , Xenopus Proteins/chemistry , Xenopus Proteins/metabolism , Y-Box-Binding Protein 1/analysis , Y-Box-Binding Protein 1/chemistry , Y-Box-Binding Protein 1/metabolism , Zyxin/analysis , Zyxin/chemistry , Zyxin/metabolism
3.
Int J Mol Sci ; 22(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33808029

ABSTRACT

Members of the human Zyxin family are LIM domain-containing proteins that perform critical cellular functions and are indispensable for cellular integrity. Despite their importance, not much is known about their structure, functions, interactions and dynamics. To provide insights into these, we used a set of in-silico tools and databases and analyzed their amino acid sequence, phylogeny, post-translational modifications, structure-dynamics, molecular interactions, and functions. Our analysis revealed that zyxin members are ohnologs. Presence of a conserved nuclear export signal composed of LxxLxL/LxxxLxL consensus sequence, as well as a possible nuclear localization signal, suggesting that Zyxin family members may have nuclear and cytoplasmic roles. The molecular modeling and structural analysis indicated that Zyxin family LIM domains share similarities with transcriptional regulators and have positively charged electrostatic patches, which may indicate that they have previously unanticipated nucleic acid binding properties. Intrinsic dynamics analysis of Lim domains suggest that only Lim1 has similar internal dynamics properties, unlike Lim2/3. Furthermore, we analyzed protein expression and mutational frequency in various malignancies, as well as mapped protein-protein interaction networks they are involved in. Overall, our comprehensive bioinformatic analysis suggests that these proteins may play important roles in mediating protein-protein and protein-nucleic acid interactions.


Subject(s)
Computational Biology , Nuclear Export Signals , Zyxin , Humans , Protein Domains , Protein Transport , Structure-Activity Relationship , Zyxin/chemistry , Zyxin/genetics , Zyxin/metabolism
4.
Mol Biol Cell ; 32(13): 1221-1228, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33909446

ABSTRACT

Sensing physical forces is a critical first step in mechano-transduction of cells. Zyxin, a LIM domain-containing protein, is recruited to force-bearing actin filaments and is thought to repair and strengthen them. Yet, the precise force-induced protein interactions surrounding zyxin remain unclear. Using BioID analysis, we identified proximal proteins surrounding zyxin under normal and force-bearing conditions by label-free mass spectrometry analysis. Under force-bearing conditions, increased biotinylation of α-actinin 1, α-actinin 4, and AFAP1 were detected, and these proteins accumulated along force-bearing actin fibers independently from zyxin, albeit at a lower intensity than zyxin. VASP also accumulated along force-bearing actin fibers in a zyxin-dependent manner, but the biotinylation of VASP remained constant regardless of force, supporting the model of a free zyxin-VASP complex in the cytoplasm being corecruited to tensed actin fibers. In addition, ARHGAP42, a RhoA GAP, was also identified as a proximal protein of zyxin and colocalized with zyxin along contractile actin bundles. The overexpression of ARHGAP42 reduced the rate of small wound closure, a zyxin-dependent process. These results demonstrate that the application of proximal biotinylation can resolve the proximity and composition of protein complexes as a function of force, which had not been possible with traditional biochemical analysis.


Subject(s)
Biomechanical Phenomena/physiology , Zyxin/metabolism , Zyxin/physiology , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Cell Adhesion Molecules/metabolism , Dogs , Focal Adhesions/metabolism , Madin Darby Canine Kidney Cells , Mechanical Phenomena , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Stress, Mechanical , Zyxin/chemistry
5.
J Phys Chem Lett ; 11(23): 10233-10241, 2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33206530

ABSTRACT

Focal adhesions (FAs) are dynamic protein nanostructures that form mechanical links between cytoskeletal actin fibers and the extracellular matrix. Here, we demonstrate that interferometric scattering (iSCAT) microscopy, a high-speed and time-unlimited imaging technique, can uncover the real-time dynamics of nanoscopic nascent adhesions (NAs). The high sensitivity and stability of the iSCAT signal enabled us to trace the whole life span of each NA spontaneously nucleated under a lamellipodium. Such high-throughput and long-term image data provide a unique opportunity for statistical analysis of adhesion dynamics. Moreover, we directly revealed that FAs play critical roles in both the extrusion of filopodia as nucleation sites on the leading edge and the one-dimensional transport of cargos along cytoskeletal fibers as fiber docking sites. These experimental results show that iSCAT is a sensitive tool for tracking real-time dynamics of nanoscopic objects involved in endogenous and exogenous biological processes in living cells.


Subject(s)
Fluorescence , Optical Imaging , Cell Adhesion , Cell Line, Tumor , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Interference , Zyxin/chemistry , Zyxin/metabolism
6.
Anticancer Res ; 40(11): 5981-5988, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33109535

ABSTRACT

Zyxin (ZYX) is a LIM domain protein whose presence has been detected in the cytoplasm and nucleus. ZYX can translocate between these two compartments and therefore, can take part in the regulation of various cellular processes. VASP and α-actinin are examples of proteins that interact with ZYX. As ZYX is present in focal adhesions (FAs), an immense part of research is focused on the role of this protein in the organisation and function of the cytoskeleton. Other studies aim to explain the impact of zyxin on other intracellular processes. Zyxin has been shown to take part in apoptosis, as well as in wound healing. Additionally, zyxin contribution to cancer development is gaining growing interest. This paper aims to systematise the knowledge on zyxin and its role in carcinogenesis.


Subject(s)
Carcinogenesis/metabolism , Zyxin/metabolism , Animals , Humans , Neoplasms/metabolism , Neoplasms/pathology , Zyxin/chemistry
7.
Biochemistry ; 56(35): 4626-4636, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28783324

ABSTRACT

Vasodilator-stimulated phosphoprotein (VASP) is a processive actin polymerase with roles in the control of cell shape and cell migration. Through interaction with the cytoskeletal adaptor protein Zyxin, VASP can localize to damaged stress fibers where it serves to repair and reinforce these structures. VASP localization is mediated by its N-terminal Ena/VASP homology (EVH1) domain, which binds to the (W/F)PxφP motif (most commonly occurring as FPPPP) found in cytoskeletal proteins such as vinculin, lamellipodin, and Zyxin. Sequentially close clusters of four or five of these motifs frequently occur, as in the proline rich region of Zyxin with four such motifs. This suggests that tetrameric VASP might bind very tightly to Zyxin through avidity, with all four EVH1 domains binding to a single Zyxin molecule. Here, quantitative nuclear magnetic resonance titration analysis reveals a dominant bivalent 1:1 (Zyxin:EVH1) interaction between the Zyxin proline rich region and the VASP EVH1 domain that utilizes the EVH1 canonical binding site and a novel secondary binding site on the opposite face of the EVH1 domain. We further show that binding to the secondary binding site is specifically inhibited by mutation of VASP EVH1 domain residue Y39 to E, which mimics Abl-induced phosphorylation of Y39. On the basis of these findings, we propose a model in which phosphorylation of Y39 acts as a stoichiometry switch that governs binding partner selection by the constitutive VASP tetramer. These results have broader implications for other multivalent VASP EVH1 domain binding partners and for furthering our understanding of the role of Y39 phosphorylation in regulating VASP localization and cellular function.


Subject(s)
Cell Adhesion Molecules/chemistry , Microfilament Proteins/chemistry , Phosphoproteins/chemistry , Zyxin/chemistry , Amino Acid Sequence , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Conformation , Protein Domains
8.
ChemMedChem ; 11(12): 1309-17, 2016 06 20.
Article in English | MEDLINE | ID: mdl-26497622

ABSTRACT

The amyloidogenic pathway is a prominent feature of Alzheimer's disease (AD). However, growing evidence suggests that a linear disease model based on ß-amyloid peptide (Aß) alone is not likely to be realistic, which therefore calls for further investigations on the other actors involved in the play. The pro-oxidant environment induced by Aß in AD pathology is well established, and a correlation among Aß, oxidative stress, and conformational changes in p53 has been suggested. In this study, we applied a multifunctional approach to identify allyl thioesters of variously substituted trans-cinnamic acids for which the pharmacological profile was strategically tuned by hydroxy substituents on the aromatic moiety. Indeed, only catechol derivative 3 [(S)-allyl (E)-3-(3,4-dihydroxyphenyl)prop-2-enethioate] inhibited Aß fibrilization. Conversely, albeit to different extents, all compounds were able to decrease the formation of reactive oxygen species in SH-SY5Y neuroblastoma cells and to prevent alterations in the conformation of p53 and its activity mediated by soluble sub-lethal concentrations of Aß. This may support an involvement of oxidative stress in Aß function, with p53 emerging as a potential mediator of their functional interplay.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Ligands , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cinnamates/chemistry , Humans , Hydrogen Peroxide/toxicity , Oxidative Stress/drug effects , Protective Agents/pharmacology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Zyxin/chemistry , Zyxin/metabolism
9.
PLoS One ; 10(10): e0140511, 2015.
Article in English | MEDLINE | ID: mdl-26509500

ABSTRACT

Focal adhesions are integrin-based structures that link the actin cytoskeleton and the extracellular matrix. They play an important role in various cellular functions such as cell signaling, cell motility and cell shape. To ensure and fine tune these different cellular functions, adhesions are regulated by a large number of proteins. The LIM domain protein zyxin localizes to focal adhesions where it participates in the regulation of the actin cytoskeleton. Because of its interactions with a variety of binding partners, zyxin has been proposed to act as a molecular scaffold. Here, we studied the interaction of zyxin with such a partner: Tes. Similar to zyxin, Tes harbors three highly conserved LIM domains of which the LIM1 domain directly interacts with zyxin. Using different zyxin variants in pull-down assays and ectopic recruitment experiments, we identified the Tes binding site in zyxin and showed that four highly conserved amino acids are crucial for its interaction with Tes. Based upon these findings, we used a zyxin mutant defective in Tes-binding to assess the functional consequences of abrogating the zyxin-Tes interaction in focal adhesions. Performing fluorescence recovery after photobleaching, we showed that zyxin recruits Tes to focal adhesions and modulates its turnover in these structures. However, we also provide evidence for zyxin-independent localization of Tes to focal adhesions. Zyxin increases focal adhesion numbers and reduces focal adhesion lifetimes, but does so independent of Tes. Quantitative analysis showed that the loss of interaction between zyxin and Tes affects the process of cell spreading. We conclude that zyxin influences focal adhesion dynamics, that it recruits Tes and that this interaction is functional in regulating cell spreading.


Subject(s)
DNA-Binding Proteins/metabolism , Protein Interaction Mapping , Zyxin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Count , Cell Movement , Conserved Sequence , Cytoskeletal Proteins , Focal Adhesions/metabolism , Humans , Kinetics , Mice , Mitochondria/metabolism , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA-Binding Proteins , Structure-Activity Relationship , Zyxin/chemistry
10.
PLoS One ; 10(4): e0120616, 2015.
Article in English | MEDLINE | ID: mdl-25860875

ABSTRACT

α-Actinins (ACTNs) are known to crosslink actin filaments at focal adhesions in migrating cells. Among the four isoforms of mammalian ACTNs, ACTN1 and ACTN4 are ubiquitously expressed. Recently, ACTN4 was reported to enhance cancer cell motility, invasion, and metastasis. However, the mechanism by which ACTN4 drives these malignant phenotypes remains unclear. Here, we show that ACTN4, but not ACTN1, induces the formation of immature focal adhesions in DLD-1 cells, leading to the rapid turnover of focal adhesions. Interestingly, zyxin (ZYX) assembly to focal adhesions was markedly decreased in ACTN4-expressing DLD-1 cells, while the recruitment of paxillin (PAX) occurred normally. On the other hand, in ACTN1-expressing DLD-1 cells, PAX and ZYX were normally recruited to focal adhesions, suggesting that ACTN4 specifically impairs focal adhesion maturation by inhibiting the recruitment of ZYX to focal complexes. Using purified recombinant proteins, we found that ZYX binding to ACTN4 was defective under conditions where ZYX binding to ACTN1 was observed. Furthermore, Matrigel invasion of SW480 cells that express high endogenous levels of ACTN4 protein was inhibited by ectopic expression of ACTN1. Altogether, our results suggest that ZYX defective binding to ACTN4, which occupies focal adhesions instead of ACTN1, induces the formation of immature focal adhesions, resulting in the enhancement of cell motility and invasion.


Subject(s)
Actinin/metabolism , Focal Adhesions/metabolism , Actinin/antagonists & inhibitors , Actinin/genetics , Caco-2 Cells , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HT29 Cells , Humans , Microscopy, Fluorescence , Neoplasm Invasiveness , Paxillin/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Zyxin/chemistry , Zyxin/genetics , Zyxin/metabolism
11.
Proc Natl Acad Sci U S A ; 112(16): 5011-6, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25848013

ABSTRACT

Small-molecule competitors of protein-protein interactions are urgently needed for functional analysis of large-scale genomics and proteomics data. Particularly abundant, yet so far undruggable, targets include domains specialized in recognizing proline-rich segments, including Src-homology 3 (SH3), WW, GYF, and Drosophila enabled (Ena)/vasodilator-stimulated phosphoprotein (VASP) homology 1 (EVH1) domains. Here, we present a modular strategy to obtain an extendable toolkit of chemical fragments (ProMs) designed to replace pairs of conserved prolines in recognition motifs. As proof-of-principle, we developed a small, selective, peptidomimetic inhibitor of Ena/VASP EVH1 domain interactions. Highly invasive MDA MB 231 breast-cancer cells treated with this ligand showed displacement of VASP from focal adhesions, as well as from the front of lamellipodia, and strongly reduced cell invasion. General applicability of our strategy is illustrated by the design of an ErbB4-derived ligand containing two ProM-1 fragments, targeting the yes-associated protein 1 (YAP1)-WW domain with a fivefold higher affinity.


Subject(s)
Proline-Rich Protein Domains , Protein Interaction Mapping , Animals , Cell Adhesion Molecules/chemistry , Cell Line, Tumor , Cell Membrane Permeability , Crystallography, X-Ray , Drosophila melanogaster/metabolism , Esterification , Fluorescent Antibody Technique , Humans , Kinetics , Ligands , Microfilament Proteins/chemistry , Models, Molecular , Molecular Weight , Peptides/chemistry , Phosphoproteins/chemistry , Protein Binding , Protein Structure, Tertiary , Pseudopodia , Stress Fibers/metabolism , Zyxin/chemistry
12.
Proc Natl Acad Sci U S A ; 111(11): 3949-54, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24594603

ABSTRACT

Cell migration through 3D extracellular matrices is critical to the normal development of tissues and organs and in disease processes, yet adequate analytical tools to characterize 3D migration are lacking. Here, we quantified the migration patterns of individual fibrosarcoma cells on 2D substrates and in 3D collagen matrices and found that 3D migration does not follow a random walk. Both 2D and 3D migration features a non-Gaussian, exponential mean cell velocity distribution, which we show is primarily a result of cell-to-cell variations. Unlike in the 2D case, 3D cell migration is anisotropic: velocity profiles display different speed and self-correlation processes in different directions, rendering the classical persistent random walk (PRW) model of cell migration inadequate. By incorporating cell heterogeneity and local anisotropy to the PRW model, we predict 3D cell motility over a wide range of matrix densities, which identifies density-independent emerging migratory properties. This analysis also reveals the unexpected robust relation between cell speed and persistence of migration over a wide range of matrix densities.


Subject(s)
Cell Movement/physiology , Extracellular Matrix , Models, Biological , Actinin/chemistry , Anisotropy , Cell Line, Tumor , Computer Simulation , Crk-Associated Substrate Protein/chemistry , Humans , Stochastic Processes , Zyxin/chemistry
13.
Biochemistry ; 53(2): 350-60, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24393048

ABSTRACT

The class XX myosin is a member of the diverse myosin superfamily and exists in insects and several lower invertebrates. DmMyo20, the class XX myosin in Drosophila, is encoded by dachs, which functions as a crucial downstream component of the Fat signaling pathway, influencing growth, affinity, and gene expression during development. Sequence analysis shows that DmMyo20 contains a unique N-terminal extension, the motor domain, followed by one IQ motif, and a C-terminal tail. To investigate the biochemical properties of DmMyo20, we expressed several DmMyo20 truncated constructs containing the motor domain in the baculovirus/Sf9 system. We found that the motor domain of DmMyo20 had neither ATPase activity nor the ability to bind to ATP, suggesting that DmMyo20 does not function as a molecular motor. We found that the motor domain of DmMyo20 could specifically bind to actin filaments in an ATP-independent manner and enhance the interaction between actin filaments and Zyx102, a downstream component of DmMyo20 in the Fat signaling pathway. These results suggest that DmMyo20 functions as a scaffold protein, but not as a molecular motor, in a signaling pathway controlling cell differentiation.


Subject(s)
Actins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microfilament Proteins/metabolism , Myosins/metabolism , Zyxin/metabolism , Actins/chemistry , Animals , Cloning, Molecular , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Myosins/chemistry , Myosins/genetics , Zyxin/chemistry
14.
PLoS One ; 8(8): e69378, 2013.
Article in English | MEDLINE | ID: mdl-23990882

ABSTRACT

Contractile actomyosin stress fibers are critical for maintaining the force balance between the interior of the cell and its environment. Consequently, the actin cytoskeleton undergoes dynamic mechanical loading. This results in spontaneous, stochastic, highly localized strain events, characterized by thinning and elongation within a discrete region of stress fiber. Previous work showed the LIM-domain adaptor protein, zyxin, is essential for repair and stabilization of these sites. Using live imaging, we show paxillin, another LIM-domain adaptor protein, is also recruited to stress fiber strain sites. Paxillin recruitment to stress fiber strain sites precedes zyxin recruitment. Zyxin and paxillin are each recruited independently of the other. In cells lacking paxillin, actin recovery is abrogated, resulting in slowed actin recovery and increased incidence of catastrophic stress fiber breaks. For both paxillin and zyxin, the LIM domains are necessary and sufficient for recruitment. This work provides further evidence of the critical role of LIM-domain proteins in responding to mechanical stress in the actin cytoskeleton.


Subject(s)
Actins/chemistry , Paxillin/chemistry , Stress Fibers/metabolism , Zyxin/chemistry , Actomyosin/metabolism , Animals , Cell Line , Cell Separation , Cytoskeleton/metabolism , Fibroblasts/metabolism , Flow Cytometry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Green Fluorescent Proteins/metabolism , Homeostasis , Image Processing, Computer-Assisted , Mice , Mice, Transgenic , Microscopy, Fluorescence , Phosphotyrosine/chemistry , Protein Structure, Tertiary , RNA Interference , Signal Transduction , Stochastic Processes
15.
Mol Biol Cell ; 24(8): 1232-49, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23427270

ABSTRACT

In vertebrates, zyxin is a LIM-domain protein belonging to a family composed of seven members. We show that the nematode Caenorhabditis elegans has a unique zyxin-like protein, ZYX-1, which is the orthologue of the vertebrate zyxin subfamily composed of zyxin, migfilin, TRIP6, and LPP. The ZYX-1 protein is expressed in the striated body-wall muscles and localizes at dense bodies/Z-discs and M-lines, as well as in the nucleus. In yeast two-hybrid assays ZYX-1 interacts with several known dense body and M-line proteins, including DEB-1 (vinculin) and ATN-1 (α-actinin). ZYX-1 is mainly localized in the middle region of the dense body/Z-disk, overlapping the apical and basal regions containing, respectively, ATN-1 and DEB-1. The localization and dynamics of ZYX-1 at dense bodies depend on the presence of ATN-1. Fluorescence recovery after photobleaching experiments revealed a high mobility of the ZYX-1 protein within muscle cells, in particular at dense bodies and M-lines, indicating a peripheral and dynamic association of ZYX-1 at these muscle adhesion structures. A portion of the ZYX-1 protein shuttles from the cytoplasm into the nucleus, suggesting a role for ZYX-1 in signal transduction. We provide evidence that the zyx-1 gene encodes two different isoforms, ZYX-1a and ZYX-1b, which exhibit different roles in dystrophin-dependent muscle degeneration occurring in a C. elegans model of Duchenne muscular dystrophy.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Dystrophin/metabolism , Muscles/metabolism , Zyxin/physiology , Actinin/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/chemistry , Gene Expression , Molecular Sequence Data , Muscles/cytology , Organ Specificity , Phylogeny , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Transport , Sequence Homology, Amino Acid , Zyxin/chemistry
16.
Cell Mol Biol Lett ; 18(1): 1-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23076992

ABSTRACT

Cell-cell and cell-substrate adhesions are sites of dramatic actin rearrangements and where actin-membrane connections are tightly regulated. Zyxin-VASP complexes localize to sites of cell-cell and cell-substrate adhesion and function to regulate actin dynamics and actin-membrane connections at these sites. To accomplish these functions, zyxin recruits VASP to cellular sites via proline-rich binding sites near zyxin's amino terminus. While the prevailing thought has been that zyxin simply acts as a scaffold protein for VASP binding, the identification of a LIM domain-VASP interaction could complicate this view. Here we assess how zyxin-VASP binding through both the proline rich motifs and the LIM domains alters specific VASP functions. We find that neither individual interaction alters VASP's actin regulatory activities. In contrast, however, we find that full-length zyxin dramatically reduces VASP-mediated actin bundling and actin assembly. Taken together, these results suggest a model where zyxin-VASP complexes occur in complex organizations with suppressed actin regulatory activity.


Subject(s)
Actins/metabolism , Cell Adhesion Molecules/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Zyxin/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/genetics , Binding Sites , Cell Adhesion , Cell Adhesion Molecules/chemistry , Cell Communication , Eukaryotic Cells/chemistry , Eukaryotic Cells/metabolism , Humans , Microfilament Proteins/chemistry , Phosphoproteins/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Zyxin/chemistry
17.
Biochem Biophys Res Commun ; 422(4): 653-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22609203

ABSTRACT

The ability to sense mechanical forces is vital to cell physiology. Yet, the molecular basis of mechano-signaling remains unclear. Previous studies have shown that zyxin, a focal adhesion protein, is recruited at force-bearing sites on the actin cytoskeleton and, therefore, identifying zyxin as a mechano-sensing protein candidate. Furthermore, zyxin accumulation at force-bearing sites requires the LIM domain located at the C-terminus of zyxin. The zyxin LIM domain consists of three LIM motifs, each containing two zinc-binding sites. Since individual LIM motifs do not accumulate at focal adhesions or force-bearing sites, we hypothesize that multiple zyxin LIM domains increase force sensitivity. Using a miniature force sensor and GFP-tagged LIM variants, we quantified the relationship between single, tandem dimer and trimer LIM protein localization and traction forces. While the presence of extra LIM domains affected VASP recruitment to focal adhesions, force sensitivity was not enhanced over the single LIM domain. Therefore, zyxin force sensitivity is optimal with a single LIM domain, while additional LIM domains fail to enhance force sensitivity.


Subject(s)
LIM Domain Proteins/metabolism , Mechanotransduction, Cellular , Zyxin/metabolism , Animals , Cell Adhesion Molecules/metabolism , Dogs , Focal Adhesions , LIM Domain Proteins/chemistry , LIM Domain Proteins/genetics , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Protein Structure, Tertiary , Zyxin/chemistry , Zyxin/genetics
18.
Biochem Biophys Res Commun ; 415(3): 485-9, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-22056563

ABSTRACT

Cell-cell junction remodeling is associated with dramatic actin reorganizations. Several actin regulatory systems have been implicated in actin remodeling events as cell-cell contacts are assembled and disassembled, including zyxin/LPP-VASP complexes. These complexes facilitate strong cell-cell adhesion by maintaining actin-membrane connections. It has been proposed that zyxin and LPP localize to cell-cell junctions via a well-defined interaction with alpha-actinin. This was recently confirmed for LPP, but zyxin localization at cell-cell contacts occurs independently of alpha-actinin binding. Here we seek to map the zyxin sequence responsible for localization to cell-cell contacts and identify the protein that docks zyxin at this cellular location. Previous results have shown that a zyxin fragment excluding the alpha-actin binding site and the LIM domains (amino acids 51-392) can independently localize to cell-cell contacts. Here, expression of smaller zyxin fragments show that zyxin localization requires amino acids 230-280. A yeast-two-hybrid screen, using the central region of zyxin as bait, resulted in the identification of the cell-cell adhesion receptor nectin-4 as a zyxin binding partner. Further demonstrating zyxin-nectin interactions, zyxin binds the intracellular domain of nectin-2 in vitro. Depletion of nectin-2 from L cells expressing E-cadherin results in a loss of zyxin localization to cell-cell contacts, demonstrating that the zyxin-nectin interaction plays a critical role in zyxin targeting to these sites.


Subject(s)
Cell Adhesion Molecules/metabolism , Zyxin/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Line , Dogs , Humans , Nectins , Protein Structure, Tertiary , Two-Hybrid System Techniques , Zyxin/chemistry , Zyxin/genetics
19.
Biophys J ; 101(5): 1069-75, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21889443

ABSTRACT

Cellular responses to mechanical perturbation are vital to cell physiology. In particular, migrating cells have been shown to sense substrate stiffness and alter cell morphology and speed. Zyxin is a focal adhesion protein that responds to external mechanical forces; however, the mechanisms of zyxin recruitment at force-bearing sites are unknown. Using force-sensing microfabricated substrates, we simultaneously measured traction force and zyxin recruitment at force-bearing sites. GFP-tagged zyxin accumulates at force-bearing sites at the leading edge, but not at the trailing edge, of migrating epithelial cells. Zyxin recruitment at force-bearing sites depends on Rho-kinase and myosin II activation, suggesting that zyxin responds not only to the externally applied force, as previously shown, but also to the internally generated actin-myosin force. Zyxin in turn recruits vasodilator-stimulated phosphoprotein, a regulator of actin assembly, to force-bearing sites. To dissect the domains of zyxin that are essential for this unique force-dependent accumulation, we generated two zyxin truncation mutants: one lacking the LIM domain (ΔLIM) and one containing only the LIM domain with all three LIM motifs (LIM). GFP-tagged ΔLIM does not localize to the force-bearing sites, but GFP-tagged zyxin LIM-domain is sufficient for the recruitment to and dynamics at force-bearing focal adhesions. Furthermore, one or two LIM motifs are not sufficient for force-dependent accumulation, suggesting that all three LIM motifs are required. Therefore, the LIM domain of zyxin recruits zyxin to force-bearing sites at the leading edge of migrating cells.


Subject(s)
Cell Movement , Mechanical Phenomena , Zyxin/chemistry , Zyxin/metabolism , Amino Acid Motifs , Animals , Binding Sites , Biomechanical Phenomena , Cell Adhesion Molecules/metabolism , Cell Line , Dogs , Humans , Microfilament Proteins/metabolism , Myosin Type II/metabolism , Phosphoproteins/metabolism , Protein Structure, Tertiary , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...