Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 300(5): 107234, 2024 May.
Article in English | MEDLINE | ID: mdl-38552737

ABSTRACT

Focal adhesions (FAs) form the junction between extracellular matrix (ECM)-bound integrins and the actin cytoskeleton and also transmit signals that regulate cell adhesion, cytoskeletal dynamics, and cell migration. While many of these signals are rooted in reversible tyrosine phosphorylation, phosphorylation of FA proteins on Ser/Thr residues is far more abundant yet its mechanisms and consequences are far less understood. The cAMP-dependent protein kinase (protein kinase A; PKA) has important roles in cell adhesion and cell migration and is both an effector and regulator of integrin-mediated adhesion to the ECM. Importantly, subcellular localization plays a critically important role in specifying PKA function. Here, we show that PKA is present in isolated FA-cytoskeleton complexes and active within FAs in live cells. Furthermore, using kinase-catalyzed biotinylation of isolated FA-cytoskeleton complexes, we identify 53 high-stringency candidate PKA substrates within FAs. From this list, we validate tensin-3 (Tns3)-a well-established molecular scaffold, regulator of cell migration, and a component of focal and fibrillar adhesions-as a novel direct substrate for PKA. These observations identify a new pathway for phospho-regulation of Tns3 and, importantly, establish a new and important niche for localized PKA signaling and thus provide a foundation for further investigation of the role of PKA in the regulation of FA dynamics and signaling.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Focal Adhesions , Tensins , Animals , Humans , Cell Adhesion , Cell Movement , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeleton/metabolism , Focal Adhesions/enzymology , Phosphorylation , Tensins/metabolism , Mice , Rats , Cell Line , Signal Transduction/genetics
2.
Proc Natl Acad Sci U S A ; 116(14): 6766-6774, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30877242

ABSTRACT

Focal adhesion kinase (FAK) is a key signaling molecule regulating cell adhesion, migration, and survival. FAK localizes into focal adhesion complexes formed at the cytoplasmic side of cell attachment to the ECM and is activated after force generation via actomyosin fibers attached to this complex. The mechanism of translating mechanical force into a biochemical signal is not understood, and it is not clear whether FAK is activated directly by force or downstream to the force signal. We use experimental and computational single-molecule force spectroscopy to probe the mechanical properties of FAK and examine whether force can trigger activation by inducing conformational changes in FAK. By comparison with an open and active mutant of FAK, we are able to assign mechanoactivation to an initial rupture event in the low-force range. This activation event occurs before FAK unfolding at forces within the native range in focal adhesions. We are also able to assign all subsequent peaks in the force landscape to partial unfolding of FAK modules. We show that binding of ATP stabilizes the kinase domain, thereby altering the unfolding hierarchy. Using all-atom molecular dynamics simulations, we identify intermediates along the unfolding pathway, which provide buffering to allow extension of FAK in focal adhesions without compromising functionality. Our findings strongly support that forces in focal adhesions applied to FAK via known interactions can induce conformational changes, which in turn, trigger focal adhesion signaling.


Subject(s)
Adenosine Triphosphate/chemistry , Avian Proteins/chemistry , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Molecular Dynamics Simulation , Protein Unfolding , Adenosine Triphosphate/metabolism , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Chickens , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Focal Adhesions/genetics , Mechanotransduction, Cellular/genetics , Protein Domains , Structure-Activity Relationship
3.
EMBO J ; 34(4): 448-65, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25471072

ABSTRACT

Paradoxically, the thymidine kinase (TK) encoded by Kaposi sarcoma-associated herpesvirus (KSHV) is an extremely inefficient nucleoside kinase, when compared to TKs from related herpesviruses. We now show that KSHV-TK, in contrast to HSV1-TK, associates with the actin cytoskeleton and induces extensive cell contraction followed by membrane blebbing. These dramatic changes in cell morphology depend on the auto-phosphorylation of tyrosines 65, 85 and 120 in the N-terminus of KSHV-TK. Phosphorylation of tyrosines 65/85 and 120 results in an interaction with Crk family proteins and the p85 regulatory subunit of PI3-Kinase, respectively. The interaction of Crk with KSHV-TK leads to tyrosine phoshorylation of this cellular adaptor. Auto-phosphorylation of KSHV-TK also induces a loss of FAK and paxillin from focal adhesions, resulting in activation of RhoA-ROCK signalling to myosin II and cell contraction. In the absence of FAK or paxillin, KSHV-TK has no effect on focal adhesion integrity or cell morphology. Our observations demonstrate that by acting as a tyrosine kinase, KSHV-TK modulates signalling and cell morphology.


Subject(s)
Focal Adhesions/enzymology , Focal Adhesions/metabolism , Herpesvirus 8, Human/enzymology , Protein-Tyrosine Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Paxillin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-crk/metabolism
4.
J Cell Physiol ; 232(9): 2447-2460, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27591737

ABSTRACT

Glaucoma, a leading cause of irreversible blindness, is commonly associated with elevated intraocular pressure (IOP) due to impaired aqueous humor (AH) drainage through the trabecular meshwork (TM). Although dysregulated production and organization of extracellular matrix (ECM) is presumed to increase resistance to AH outflow and elevate IOP by altering TM cell contractile and adhesive properties, it is not known whether regulation of ECM protein phosphorylation via the secretory vertebrate lonesome kinase (VLK) influences TM cellular characteristics. Here, we tested this possibility. Experiments carried out in this study reveal that the 32 kDa protein is a prominent VLK isoform detectable in lysates and conditioned media (CM) of human TM cells. Increased levels of VLK were observed in CM of TM cells subjected to cyclic mechanical stretch, or treated with dexamethasone, TGF-ß2, and TM cells expressing constitutively active RhoA GTPase. Downregulation of VLK expression in TM cells using siRNA decreased tyrosine phosphorylation (TyrP) of ECM proteins and focal adhesions, and induced changes in cell shape in association with reduced levels of actin stress fibers and phospho-paxillin. VLK was also demonstrated to regulate TGF-ß2-induced TyrP of ECM proteins. Taken together, these results suggest that VLK secretion can be regulated by external cues, intracellular signal proteins, and mechanical stretch, and VLK can in turn regulate TyrP of ECM proteins secreted by TM cells and control cell shape, actin stress fibers, and focal adhesions. These observations indicate a potential role for VLK in homeostasis of AH outflow and IOP, and in the pathobiology of glaucoma. J. Cell. Physiol. 232: 2447-2460, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Adhesion , Cell Shape , Extracellular Matrix Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Trabecular Meshwork/enzymology , Adult , Aged , Aqueous Humor/metabolism , Cell Adhesion/drug effects , Cell Shape/drug effects , Cells, Cultured , Culture Media, Conditioned/metabolism , Dexamethasone/pharmacology , Focal Adhesions/enzymology , Glaucoma/enzymology , Glaucoma/pathology , Glaucoma/physiopathology , Humans , Intraocular Pressure , Mechanotransduction, Cellular , Middle Aged , Mutation , Paxillin/metabolism , Phosphorylation , Protein-Tyrosine Kinases/genetics , RNA Interference , Stress Fibers/enzymology , Time Factors , Trabecular Meshwork/drug effects , Transfection , Transforming Growth Factor beta2/pharmacology , Tyrosine , Young Adult , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
5.
J Biol Chem ; 289(26): 18427-41, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24821720

ABSTRACT

Interleukin-1 (IL-1) signaling in fibroblasts is mediated through focal adhesions, organelles that are enriched with adaptor and cytoskeletal proteins that regulate signal transduction. We examined interactions of the focal adhesion kinase (FAK) with protein-tyrosine phosphatase-α (PTP-α) in IL-1 signaling. In wild type and FAK knock-out mouse embryonic fibroblasts, we found by immunoblotting, immunoprecipitation, immunostaining, and gene silencing that FAK is required for IL-1-mediated sequestration of PTPα to focal adhesions. Immunoprecipitation and pulldown assays of purified proteins demonstrated a direct interaction between FAK and PTPα, which was dependent on the FAT domain of FAK and by an intact membrane-proximal phosphatase domain of PTPα. Recruitment of PTPα to focal adhesions, IL-1-induced Ca(2+) release from the endoplasmic reticulum, ERK activation, and IL-6, MMP-3, and MMP-9 expression were all blocked in FAK knock-out fibroblasts. These processes were restored in FAK knock-out cells transfected with wild type FAK, FAT domain, and FRNK. Our data indicate that IL-1-induced signaling through focal adhesions involves interactions between the FAT domain of FAK and PTPα.


Subject(s)
Fibroblasts/enzymology , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Interleukin-1/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesions/metabolism , Interleukin-1/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , NIH 3T3 Cells , Protein Binding , Protein Structure, Tertiary , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Signal Transduction
6.
Biochem Biophys Res Commun ; 464(1): 275-80, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26116771

ABSTRACT

Cancer cell invasion is a critical phenomenon in cancer pathogenesis. Glycogen synthase kinase-3ß (GSK-3ß) has been reported to regulate cancer cell invasion both negatively and positively. Thus, the net effect of GSK-3ß on invasion is unclear. In this report, we showed that GSK-3ß inhibitors induced dysregulation of the actin cytoskeleton and functional insufficiency of focal adhesion, which resulted in suppressed invasion. In addition, WAVE2, an essential molecule for actin fibre branching, was down-regulated after GSK-3ß inhibition. Collectively, we propose that the WAVE2-actin cytoskeleton axis is an important target of GSK-3ß inhibitors in cancer cell invasion.


Subject(s)
Actin Cytoskeleton/drug effects , Cell Movement/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , Urea/analogs & derivatives , Wiskott-Aldrich Syndrome Protein Family/antagonists & inhibitors , Actin Cytoskeleton/enzymology , Actin Cytoskeleton/ultrastructure , Actins/chemistry , Actins/genetics , Actins/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Collagen/chemistry , Drug Combinations , Focal Adhesions/drug effects , Focal Adhesions/enzymology , Focal Adhesions/ultrastructure , Gene Expression , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Laminin/chemistry , Proteoglycans/chemistry , Urea/pharmacology , Wiskott-Aldrich Syndrome Protein Family/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism
7.
Am J Pathol ; 184(10): 2742-56, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25239564

ABSTRACT

Aberrant focal adhesion turnover is centrally involved in podocyte actin cytoskeleton disorganization and foot process effacement. The structural and dynamic integrity of focal adhesions is orchestrated by multiple cell signaling molecules, including glycogen synthase kinase 3ß (GSK3ß), a multitasking kinase lately identified as a mediator of kidney injury. However, the role of GSK3ß in podocytopathy remains obscure. In doxorubicin (Adriamycin)-injured podocytes, lithium, a GSK3ß inhibitor and neuroprotective mood stabilizer, obliterated the accelerated focal adhesion turnover, rectified podocyte hypermotility, and restored actin cytoskeleton integrity. Mechanistically, lithium counteracted the doxorubicin-elicited GSK3ß overactivity and the hyperphosphorylation and overactivation of paxillin, a focal adhesion-associated adaptor protein. Moreover, forced expression of a dominant negative kinase dead mutant of GSK3ß highly mimicked, whereas ectopic expression of a constitutively active GSK3ß mutant abolished, the effect of lithium in doxorubicin-injured podocytes, suggesting that the effect of lithium is mediated, at least in part, through inhibition of GSK3ß. Furthermore, paxillin interacted with GSK3ß and served as its substrate. In mice with doxorubicin nephropathy, a single low dose of lithium ameliorated proteinuria and glomerulosclerosis. Consistently, lithium therapy abrogated GSK3ß overactivity, blunted paxillin hyperphosphorylation, and reinstated actin cytoskeleton integrity in glomeruli associated with an early attenuation of podocyte foot process effacement. Thus, GSK3ß-modulated focal adhesion dynamics might serve as a novel therapeutic target for podocytopathy.


Subject(s)
Focal Adhesions/enzymology , Glycogen Synthase Kinase 3/metabolism , Kidney Glomerulus/enzymology , Lithium/pharmacology , Paxillin/metabolism , Podocytes/physiology , Actin Cytoskeleton/drug effects , Amino Acid Sequence , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Doxorubicin/pharmacology , Focal Adhesions/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Kidney Glomerulus/drug effects , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phosphorylation/drug effects , Podocytes/drug effects , Podocytes/enzymology , Podocytes/pathology , Signal Transduction/drug effects
8.
Curr Opin Cell Biol ; 20(1): 85-90, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18083360

ABSTRACT

Cell migration requires the integration and coordination of specific focal adhesion dynamics at the cell front, center and rear. In this review, we will present our understanding of the regulation of adhesion turnover and disassembly in various regions of the cell. Adhesion turnover involves a number of tyrosine kinases and phosphatases, most of which are engaged in FAK signaling pathways. Additionally, adhesions are regulated by tensile forces that depend on dynamic coupling with the actin cytoskeleton. The distribution of adhesion disassembly throughout a motile cell is likely coordinated by the asymmetry of the microtubule network. We present a model that suggests two stages of microtubule-driven adhesion disassembly: destabilization and detachment.


Subject(s)
Cell Movement , Cell Polarity , Focal Adhesions/metabolism , Actins/metabolism , Animals , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Humans , Microtubules/metabolism
9.
Nat Cell Biol ; 10(9): 1017-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18758489

ABSTRACT

Talin can activate integrins to bind the extracellular matrix and also connect matrix-engaged integrins to the actin cytoskeleton. New work shows that cell spreading can be dissected into three distinct phases according to their differential requirements for talin function.


Subject(s)
Cell Movement , Cell Size , Talin/deficiency , Animals , Cell Movement/drug effects , Cell Shape/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibronectins/pharmacology , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/drug effects , Focal Adhesions/enzymology , Humans , Integrin beta1/metabolism , Mice
10.
Nat Cell Biol ; 10(9): 1062-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19160486

ABSTRACT

Cell spreading, adhesion and remodelling of the extracellular matrix (ECM) involve bi-directional signalling and physical linkages between the ECM, integrins and the cell cytoskeleton. The actin-binding proteins talin1 and 2 link ligand-bound integrins to the actin cytoskeleton and increase the affinity of integrin for the ECM. Here we report that depletion of talin2 in talin1-null (talin1(-/-)) cells did not affect the initiation of matrix-activated spreading or Src family kinase (SFK) activation, but abolished the ECM-integrin-cytoskeleton linkage and sustained cell spreading and adhesion. Specifically, focal adhesion assembly, focal adhesion kinase (FAK) signalling and traction force generation on substrates were severely affected. The talin1 head domain restored beta1 integrin activation but only full-length talin1 restored the ECM-cytoskeleton linkage and normal cytoskeleton organization. Our results demonstrate three biochemically distinct steps in fibronectin-activated cell spreading and adhesion: (1) fibronectin-integrin binding and initiation of spreading, (2) fast cell spreading and (3) focal adhesion formation and substrate traction. We suggest that talin is not required for initial cell spreading. However, talin provides the important mechanical linkage between ligand-bound integrins and the actin cytoskeleton required to catalyse focal adhesion-dependent pathways.


Subject(s)
Cell Movement , Fibroblasts/cytology , Integrin beta1/metabolism , Talin/deficiency , Actomyosin/metabolism , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Shape/drug effects , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibronectins/pharmacology , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/drug effects , Focal Adhesions/enzymology , Green Fluorescent Proteins/metabolism , Humans , Mice , Microtubules/drug effects , Microtubules/metabolism , Phosphotyrosine/metabolism , Recombinant Fusion Proteins/metabolism
12.
J Cell Sci ; 125(Pt 19): 4498-506, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22767508

ABSTRACT

Focal adhesions (FAs) are specialized membrane-associated multi-protein complexes that link the cell to the extracellular matrix and enable cell proliferation, survival and motility. Despite the extensive description of the molecular composition of FAs, the complex regulation of FA dynamics is unclear. We have used photobleaching assays of whole cells to determine the protein dynamics in every single focal adhesion. We identified that the focal adhesion proteins FAK and paxillin exist in two different states: a diffuse cytoplasmic pool and a transiently immobile FA-bound fraction with variable residence times. Interestingly, the average residence time of both proteins increased with focal adhesion size. Moreover, increasing integrin clustering by modulating surface collagen density increased residence time of FAK but not paxillin. Finally, this approach was applied to measure FAK and paxillin dynamics using nocodazole treatment followed by washout. This revealed an opposite residence time of FAK and paxillin in maturing and disassembling FAs, which depends on the ventral and peripheral cellular position of the FAs.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Paxillin/metabolism , Animals , Cell Survival/drug effects , Computer Simulation , Cytosol/drug effects , Cytosol/metabolism , Diffusion , Epithelial Cells/drug effects , Fluorescence Recovery After Photobleaching , Focal Adhesions/drug effects , Green Fluorescent Proteins/metabolism , Kinetics , LLC-PK1 Cells , Ligands , Models, Biological , Monte Carlo Method , Nocodazole/pharmacology , Protein Binding/drug effects , Recombinant Fusion Proteins/metabolism , Swine , Time Factors
13.
J Cell Sci ; 124(Pt 8): 1195-205, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21444750

ABSTRACT

The notion that cell shape and spreading can regulate cell proliferation has evolved over several years, but only recently has this been linked to forces from within and upon the cell. This emerging area of mechanical signaling is proving to be wide-spread and important for all cell types. The microenvironment that surrounds cells provides a complex spectrum of different, simultaneously active, biochemical, structural and mechanical stimuli. In this milieu, cells probe the stiffness of their microenvironment by pulling on the extracellular matrix (ECM) and/or adjacent cells. This process is dependent on transcellular cell-ECM or cell-cell adhesions, as well as cell contractility mediated by Rho GTPases, to provide a functional linkage through which forces are transmitted through the cytoskeleton by intracellular force-generating proteins. This Commentary covers recent advances in the underlying mechanisms that control cell proliferation by mechanical signaling, with an emphasis on the role of 3D microenvironments and in vivo extracellular matrices. Moreover, as there is much recent interest in the tumor-stromal interaction, we will pay particular attention to exciting new data describing the role of mechanical signaling in the progression of breast cancer.


Subject(s)
Cell Proliferation , Cytoskeleton/metabolism , Focal Adhesions/enzymology , Focal Adhesions/metabolism , Mechanotransduction, Cellular , Neoplasms/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cytoskeleton/enzymology , Cytoskeleton/genetics , Humans , Neoplasms/enzymology , Neoplasms/genetics , Signal Transduction , rho GTP-Binding Proteins/genetics
14.
Microcirculation ; 20(7): 637-49, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23600470

ABSTRACT

OBJECTIVE: To determine the role of FAK in the regulation of endothelial barrier function. METHODS: Stable FAK knockdown HLEC were generated by lentiviral infection of FAK shRNA. Measurements of isometric tension and transendothelial electrical resistance were performed. RESULTS: A FAK knockdown human pulmonary endothelial cell line was generated by lentiviral infection with FAK shRNA and resulted in greater than 90% reduction in FAK protein with no change in Pyk2 protein. Loss of FAK altered cell morphology and actin distribution in both pre- and post-confluent endothelial cells. Large, polygonal shaped endothelial cells with randomly organized stress fibers were identified in pre-confluent cultures, while in confluent monolayers, endothelial cells were irregularly shaped with actin bundles present at cell margins. An increase in the number and size of vinculin plaques was detected in FAK-depleted cells. FAK knockdown monolayers generated a greater transendothelial electrical resistance than controls. Thrombin treatment induced similar changes in TER in both FAK knockdown and control cell lines. FAK-depleted endothelial cells developed a higher stable basal isometric tension compared to control monolayers, but the increase in tension stimulated by thrombin does not differ between the cell lines. Basal myosin II regulatory light chain phosphorylation was unaltered in FAK-depleted cells. In addition, loss of FAK enhanced VE-cadherin localization to the cell membrane without altering VE-cadherin protein levels. CONCLUSIONS: The loss of FAK in endothelial cells enhanced cell attachment and strengthened cell-cell contacts resulting in greater basal tension leading to formation of a tighter endothelial monolayer.


Subject(s)
Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/enzymology , Lung/enzymology , Animals , Cell Line, Transformed , Electric Impedance , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Focal Adhesion Kinase 1/genetics , Focal Adhesions/genetics , Gene Knockdown Techniques , Humans , Lung/cytology , Mice , Mice, Knockout
15.
Mod Pathol ; 26(3): 327-35, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22996377

ABSTRACT

Ribosomal protein S6 is a key regulator of 40S ribosome biogenesis, and its phosphorylation is closely related to cell growth capacity. However, as a downstream target of S6 kinases, the clinical significance and the roles of S6 and S6 phosphorylation in cell viability and motility of esophageal squamous cell carcinoma remain unclear. Here, we show that high level of phosphorylated-ribosomal protein S6 (p-S6) (immunohistochemistry score ≥5) and an increased ratio of p-S6/S6 (immunohistochemistry score ≥0.75) were significantly associated with shortened disease-free survival in patients with esophageal squamous cell carcinoma in univariate analysis (P=0.049 and P<0.001, respectively). After adjusting for age, tumor-nodes-metastasis stage, chemotherapy, and radiation therapy in multivariate analysis, both p-S6 (hazard ratio 2.21, P=0.005) and p-S6/S6 (hazard ratio 2.40, P<0.001) remained independent adverse prognostic factors. In addition, S6 and S6 kinase 1 knockdown resulted in attenuation of viability by suppressing cyclin D1 expression in esophageal cancer cells. Furthermore, depletion of S6 and S6 kinase 1 resulted in a reduction in esophageal cancer cell migration and invasion. This was paralleled by a reduction in focal adhesion and by suppression of extracellular signal-regulated kinase and c-jun N-terminal kinase phosphorylation, which control cell motility. Collectively, these findings suggest that p-S6 and the ratio of p-S6/S6 are closely relevant to tumor progression and have prognostic significance in esophageal squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Esophageal Neoplasms/enzymology , Ribosomal Protein S6/metabolism , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cell Line, Tumor , Cell Movement , Cell Survival , Cyclin D1/metabolism , Disease-Free Survival , Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Focal Adhesions/enzymology , Humans , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases/metabolism , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Phosphorylation , Prognosis , Proportional Hazards Models , RNA Interference , Ribosomal Protein S6/genetics , Ribosomal Protein S6 Kinases/metabolism , Risk Assessment , Risk Factors , Time Factors , Transfection
16.
Biophys J ; 103(2): 175-84, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22853894

ABSTRACT

The interplay between cadherin- and integrin-dependent signals controls cell behavior, but the precise mechanisms that regulate the strength of adhesion to the extracellular matrix remains poorly understood. We deposited cells expressing a defined repertoire of cadherins and integrins on fibronectin (FN)-coated polyacrylamide gels (FN-PAG) and on FN-coated pillars used as a micro-force sensor array (µFSA), and analyzed the functional relationship between these adhesion receptors to determine how it regulates cell traction force. We found that cadherin-mediated adhesion stimulated cell spreading on FN-PAG, and this was modulated by the substrate stiffness. We compared S180 cells with cells stably expressing different cadherins on µFSA and found that traction forces were stronger in cells expressing cadherins than in parental cells. E-cadherin-mediated contact and mechanical coupling between cells are required for this increase in cell-FN traction force, which was not observed in isolated cells, and required Src and PI3K activities. Traction forces were stronger in cells expressing type I cadherins than in cells expressing type II cadherins, which correlates with our previous observation of a higher intercellular adhesion strength developed by type I compared with type II cadherins. Our results reveal one of the mechanisms whereby molecular cross talk between cadherins and integrins upregulates traction forces at cell-FN adhesion sites, and thus provide additional insight into the molecular control of cell behavior.


Subject(s)
Cadherins/metabolism , Focal Adhesions/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , src-Family Kinases/metabolism , Actins/metabolism , Animals , Biomechanical Phenomena/drug effects , Cattle , Cell Adhesion/drug effects , Cell Aggregation/drug effects , Cell Line , Cell Movement/drug effects , Chickens , Cluster Analysis , Fibronectins/pharmacology , Focal Adhesions/drug effects , Integrins/metabolism , Models, Biological , Phosphoinositide-3 Kinase Inhibitors , Protein Transport/drug effects , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors
17.
J Cell Sci ; 123(Pt 16): 2725-32, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20647370

ABSTRACT

Members of the PKC superfamily have been implicated in various migratory models and in particular in spatially restricted processes. However, defining the precise local events that underlie the PKC-dependent processes is constrained by the unspecific nature of interventions. Here we address this problem in relation to atypical PKC (aPKC) action, which in conjunction with the exocyst complex controls the polarised delivery of promigratory signals. A drug-dependent recruitment approach was employed to manipulate the local recruitment of signals to the leading edge of migrating cells, under conditions where the aPKC-exocyst control is globally abrogated. We found that activation of ERK but not JNK at focal adhesions recovers the majority of the migratory loss attributed to ERK action, demonstrating a necessary role for active plasma membrane ERK in the downstream signalling of aPKC-dependent migration. The data further show that restored focal adhesion dynamics are a contributing mechanism through which localized ERK activity influences this aPKC-exocyst-dependent migration.


Subject(s)
Cell Movement/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Protein Kinase C/metabolism , Animals , Cell Membrane/enzymology , Cells, Cultured , Enzyme Activation , Focal Adhesions/enzymology , Kidney/cytology , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Paxillin/metabolism , Phosphorylation , Protein Binding , Rats , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology
18.
Biochem Biophys Res Commun ; 417(2): 824-9, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22202174

ABSTRACT

Membrane-type matrix metalloproteinase-1 (MT1-MMP) is expressed by mechanosensitive osteocytes and affects bone mass. The extracellular domain of MT1-MMP is connected to extracellular matrix, while its intracellular domain is a strong modulator of cell signaling. In theory MT1-MMP could thus transduce mechanical stimuli into a chemical response. We hypothesized that MT1-MMP plays a role in the osteocyte response to mechanical stimuli. MT1-MMP-positive and knockdown (siRNA) MLO-Y4 osteocytes were mechanically stimulated with a pulsating fluid flow (PFF). Focal adhesions were visualized by paxillin immunostaining. Osteocyte number, number of empty lacunae, and osteocyte morphology were measured in long bones of MT1-MMP(+/+) and MT1-MMP(-/-) mice. PFF decreased MT1-MMP mRNA and protein expression in MLO-Y4 osteocytes, suggesting that mechanical loading may affect pericellular matrix remodeling by osteocytes. MT1-MMP knockdown enhanced NO production and c-jun and c-fos mRNA expression in response to PFF, concomitantly with an increased number and size of focal adhesions, indicating that MT1-MMP knockdown osteocytes have an increased sensitivity to mechanical loading. Osteocytes in MT1-MMP(-/-) bone were more elongated and followed the principle loading direction, suggesting that they might sense mechanical loading. This was supported by a lower number of empty lacunae in MT1-MMP(-/-) bone, as osteocytes lacking mechanical stimuli tend to undergo apoptosis. In conclusion, mechanical stimulation decreased MT1-MMP expression by MLO-Y4 osteocytes, and MT1-MMP knockdown increased the osteocyte response to mechanical stimulation, demonstrating a novel and unexpected role for MT1-MMP in mechanosensing.


Subject(s)
Matrix Metalloproteinase 14/physiology , Mechanotransduction, Cellular/physiology , Osteocytes/physiology , Animals , Focal Adhesions/enzymology , Focal Adhesions/genetics , Focal Adhesions/physiology , Gene Knockdown Techniques , Matrix Metalloproteinase 14/genetics , Mechanotransduction, Cellular/genetics , Mice , Mice, Mutant Strains , Osteocytes/cytology , Osteocytes/enzymology , Pulsatile Flow , RNA, Small Interfering/genetics , Stress, Mechanical
19.
Microvasc Res ; 83(1): 12-21, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21549132

ABSTRACT

Disruption of either intercellular or extracellular junctions involved in maintaining endothelial barrier function can result in increased endothelial permeability. Increased endothelial permeability, in turn, allows for the unregulated movement of fluid and solutes out of the vasculature and into the surrounding connective tissue, contributing to a number of disease states, including stroke and pulmonary edema (Ermert et al., 1995; Lee and Slutsky, 2010; van Hinsbergh, 1997; Waller et al., 1996; Warboys et al., 2010). Thus, a better understanding of the molecular mechanisms by which endothelial cell junction integrity is controlled is necessary for development of therapies aimed at treating such conditions. In this review, we will discuss the functions of three signaling molecules known to be involved in regulation of endothelial permeability: focal adhesion kinase (FAK), protein kinase C delta (PKCδ), and p190RhoGAP (p190). We will discuss the independent functions of each protein, as well as the interplay that exists between them and the effects of such interactions on endothelial function.


Subject(s)
Capillary Permeability , Endothelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Protein Kinase C-delta/metabolism , Animals , Humans , Signal Transduction
20.
Microvasc Res ; 83(1): 22-30, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21925517

ABSTRACT

The vascular endothelium serves as a semi-selective barrier between the circulating contents of the blood and the tissues through which they flow. Disruption of this barrier results in significant organ dysfunction during devastating inflammatory syndromes such as sepsis and acute lung injury (ALI). Sphingosine 1-phosphate (S1P) is an endogenous lipid regulator of endothelial permeability that produces potent barrier enhancement via actin and junctional protein rearrangement and resultant cytoskeletal changes. A key effector protein in this S1P response is focal adhesion kinase (FAK), a highly conserved cytoplasmic tyrosine kinase involved in the engagement of integrins and assembly of focal adhesions (FA) through the catalysis of multiple downstream signals. After stimulation by S1P, endothelial FAK undergoes specific tyrosine phosphorylation that results in activation of the kinase and dynamic interactions with other effector molecules to improve the endothelial barrier. FAK participates in peripheral actin cytoskeletal rearrangement as well as cell-matrix (FA) and cell-cell (adherens junction) junctional complex strengthening that combine to decrease vascular permeability. This review summarizes the current knowledge of the role of FAK in mediating enhanced endothelial barrier function by S1P.


Subject(s)
Capillary Permeability , Endothelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Enzyme Activation , Humans , Phosphorylation , Signal Transduction , Sphingosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL