20 research outputs found

    Focal adhesions: A nexus for intracellular signaling and cytoskeletal dynamics

    Get PDF
    Focal adhesions (FAs) are specialized sites of cell attachment to the extracellular matrix (ECM) where integrin receptors link the ECM to the actin cytoskeleton. Integrins cluster into supramolecular complexes with structural, cytoskeletal proteins like talin, vinculin, and a-actinin, as well as numerous signaling molecules, including c-Src, FAK, p130cas, and paxillin. The composition and molecular architecture of FAs have been reviewed elsewhere and are beyond the scope of this brief review. FAs serve at least two significant cellular functions: to transmit force or tension at adhesion sites to maintain strong attachments to the underlying ECM and to act as signaling centers from which numerous intracellular pathways emanate to regulate cell growth, survival, and gene expression. FAs are dynamic structures that assemble, disperse, and recycle (turnover) as cells migrate or enter into mitosis. Recent evidence reveals the complexity of these processes. Assembly/disassembly involves the coordinate regulation of Rho family GTPases through cross talk between integrins and numerous adhesion receptors (cadherins, cell adhesion molecules (CAMs), selectins, and syndecans), G-protein-coupled receptors (GPCRs), and receptor tyrosine kinases (RTKs), as well as the interplay between microtubules and actin. It is also apparent that FAs are themselves motile and heterogeneous in composition. Finally, turnover of FAs entails communication with components of vesicle trafficking pathways and microtubules. This review highlights recent findings relating to FA assembly, dynamics, and turnover

    Regulation of Cell Adhesion by Protein-tyrosine Phosphatases: I. CELL-MATRIX ADHESION

    Get PDF
    Cell-cell adhesion is critical to the development and maintenance of multicellular organisms. The stability of many adhesions is regulated by protein tyrosine phosphorylation of cell adhesion molecules and their associated components, with high levels of phosphorylation promoting disassembly. The level of tyrosine phosphorylation reflects the balance between protein-tyrosine kinase and protein-tyrosine phosphatase activity. Many protein-tyrosine phosphatases associate with the cadherin-catenin complex, directly regulating the phosphorylation of these proteins, thereby affecting their interactions and the integrity of cell-cell junctions. Tyrosine phosphatases can also affect cell-cell adhesions indirectly by regulating the signaling pathways that control the activities of Rho family G proteins. In addition, receptor-type tyrosine phosphatases can mediate outside-in signaling through both ligand binding and dimerization of their extracellular domains. This review will discuss the role of protein-tyrosine phosphatases in cell-cell interactions, with an emphasis on cadherin-mediated adhesions

    PTP-PEST controls motility through regulation of Rac1

    Get PDF
    The cytoplasmic protein tyrosine phosphatase, PTP-PEST, associates with the focal adhesion proteins p130cas and paxillin and has recently been implicated in cell migration. In this study, we investigated the mechanism by which PTP-PEST regulates this phenomenon. We find that PTP-PEST is activated in an adhesion-dependent manner and localizes to the tips of membrane protrusions in spreading fibroblasts. We show that the catalytic activity of PTP-PEST is a key determinant for its effects on motility. Overexpression of PTP-PEST, but not a catalytically inactive form, impairs haptotaxis, cell spreading and formation of membrane protrusions in CHOK1 cells. In addition, overexpression of PTP-PEST in Rat1 fibroblasts perturbs membrane ruffling and motility in response to PDGF stimulation. The expression level of PTP-PEST modulates the activity of the small GTPase, Rac1. PTP-PEST overexpression suppresses activation of Rac1 in response to both integrin-mediated adhesion or growth factor stimulation. In contrast, fibroblasts that lack PTP-PEST expression show enhanced Rac1 activity. Co-expression of constitutively active Rac1 with PTP-PEST overcomes the inhibition of cell spreading and migration indicating that PTP-PEST acts by antagonizing Rac1 activation. Our data suggest a model in which PTP-PEST is activated by integrins and localized to regions where it can control motile events at the leading edge through inhibition of the small GTPase Rac1

    Identification of a Filamin Docking Site on PTP-PEST

    Get PDF
    PTP-PEST is a cytoplasmic protein-tyrosine phosphatase (PTP) implicated in the regulation of biological processes such as cell motility, cytokinesis, focal adhesion disassembly, and lymphocyte activation. Using a proteomics approach, filamin-A was identified as a novel interacting protein that bound to GST-PTP-PEST. This interaction was confirmed in vitro and in cells by coimmunoprecipitation. The site of filamin interaction on PTP-PEST was mapped to the fourth proline-rich region (Pro4). PTP-PEST has previously been implicated in the regulation of cytokinesis. In further support of this finding, expression of PTP-PEST in HeLa cells resulted in the formation of multinucleated cells. A PTP-PEST mutant lacking Pro4 and unable to bind filamin-A failed to induce the multinucleated phenotype. Further, depletion of filamin-A in HeLa cells was found to reduce the PTP-PEST-dependent multinucleation phenotype. Hence, we conclude that the interaction of PTP-PEST with filamin-A may function in the control of cytokinesis in mammalian cells

    PTP-PEST Couples Membrane Protrusion and Tail Retraction via VAV2 and p190RhoGAP

    Get PDF
    Cell motility is regulated by a balance between forward protrusion and tail retraction. These phenomena are controlled by a spatial asymmetry in signals at the front and the back of the cell. We show here that the protein-tyrosine phosphatase, PTP-PEST, is required for the coupling of protrusion and retraction during cell migration. PTP-PEST null fibroblasts, which are blocked in migration, exhibit exaggerated protrusions at the leading edge and long, unretracted tails in the rear. This altered morphology is accompanied by changes in the activity of Rho GTPases, Rac1 and RhoA, which mediate protrusion and retraction, respectively. PTP-PEST null cells exhibit enhanced Rac1 activity and decreased RhoA activity. We further show that PTP-PEST directly targets the upstream regulators of Rac1 and RhoA, VAV2 and p190RhoGAP. Moreover, we demonstrate that the activities of VAV2 and p190RhoGAP are regulated by PTP-PEST. Finally, we present evidence indicating the VAV2 can be regulated by integrin-mediated adhesion. These data suggest that PTP-PEST couples protrusion and retraction by acting on VAV2 and p190RhoGAP to reciprocally modulate the activity of Rac1 and RhoA

    Integrin-mediated Adhesion Regulates Cell Polarity and Membrane Protrusion through the Rho Family of GTPases

    Get PDF
    Integrin-mediated adhesion is a critical regulator of cell migration. Here we demonstrate that integrin-mediated adhesion to high fibronectin concentrations induces a stop signal for cell migration by inhibiting cell polarization and protrusion. On fibronectin, the stop signal is generated through α5β1 integrin-mediated signaling to the Rho family of GTPases. Specifically, Cdc42 and Rac1 activation exhibits a biphasic dependence on fibronectin concentration that parallels optimum cell polarization and protrusion. In contrast, RhoA activity increases with increasing substratum concentration. We find that cross talk between Cdc42 and Rac1 is required for substratum-stimulated protrusion, whereas RhoA activity is inhibitory. We also show that Cdc42 activity is inhibited by Rac1 activation, suggesting that Rac1 activity may down-regulate Cdc42 activity and promote the formation of stabilized rather than transient protrusion. Furthermore, expression of RhoA down-regulates Cdc42 and Rac1 activity, providing a mechanism whereby RhoA may inhibit cell polarization and protrusion. These findings implicate adhesion-dependent signaling as a mechanism to stop cell migration by regulating cell polarity and protrusion via the Rho family of GTPases

    PTP-PEST controls motility, adherens junction assembly, and Rho GTPase activity in colon cancer cells

    No full text
    An important step in carcinoma progression is loss of cell-cell adhesion leading to increased invasion and metastasis. We show here that the protein tyrosine phosphatase, PTP-PEST, is a critical regulator of cell-cell junction integrity and epithelial cell motility. Using colon carcinoma cells, we show that the expression level of PTP-PEST regulates cell motility. Either transient small interfering RNA or stable short hairpin RNA knockdown of PTP-PEST enhances haptotactic and chemotactic migration of KM12C colon carcinoma cells. Furthermore, KM12C cells with stably knocked down PTP-PEST exhibit a mesenchymal-like phenotype with prominent membrane ruffles and lamellae. In contrast, ectopic expression of PTP-PEST in KM20 or DLD-1 cells, which lack detectable endogenous PTP-PEST expression, suppresses haptotactic migration. Importantly, we find that PTP-PEST localizes in adherens junctions. Concomitant with enhanced motility, stable knockdown of PTP-PEST causes a disruption of cell-cell junctions. These effects are due to a defect in junctional assembly and not to a loss of E-cadherin expression. Adherens junction assembly is impaired following calcium switch in KM12C cells with stably knocked down PTP-PEST and is accompanied by an increase in the activity of Rac1 and a suppression of RhoA activity in response to cadherin engagement. Taken together, these results suggest that PTP-PEST functions as a suppressor of epithelial cell motility by controlling Rho GTPase activity and the assembly of adherens junctions
    corecore