Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 6: 29003, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27357373

ABSTRACT

Changes in vascular permeability occur during inflammation and the actin cytoskeleton plays a crucial role in regulating endothelial cell contacts and permeability. We demonstrated recently that the actin-binding protein cortactin regulates vascular permeability via Rap1. However, it is unknown if the actin cytoskeleton contributes to increased vascular permeability without cortactin. As we consistently observed more actin fibres in cortactin-depleted endothelial cells, we hypothesised that cortactin depletion results in increased stress fibre contractility and endothelial barrier destabilisation. Analysing the contractile machinery, we found increased ROCK1 protein levels in cortactin-depleted endothelium. Concomitantly, myosin light chain phosphorylation was increased while cofilin, mDia and ERM were unaffected. Secretion of the barrier-stabilising hormone adrenomedullin, which activates Rap1 and counteracts actomyosin contractility, was reduced in plasma from cortactin-deficient mice and in supernatants of cortactin-depleted endothelium. Importantly, adrenomedullin administration and ROCK1 inhibition reduced actomyosin contractility and rescued the effect on permeability provoked by cortactin deficiency in vitro and in vivo. Our data suggest a new role for cortactin in controlling actomyosin contractility with consequences for endothelial barrier integrity.


Subject(s)
Adrenomedullin/metabolism , Capillary Permeability/physiology , Cortactin/deficiency , Endothelial Cells/physiology , Actomyosin/physiology , Animals , Contractile Proteins/biosynthesis , Contractile Proteins/genetics , Cortactin/antagonists & inhibitors , Cortactin/genetics , Cortactin/physiology , Human Umbilical Vein Endothelial Cells , Humans , Lung/cytology , Male , Mice , RNA Interference , RNA, Small Interfering/genetics , Shelterin Complex , Telomere-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/physiology
3.
PLoS One ; 6(5): e19942, 2011.
Article in English | MEDLINE | ID: mdl-21625493

ABSTRACT

Early endosomes (EEs) are known to be a sorting station for internalized molecules destined for degradation, recycling, or other intracellular organelles. Segregation is an essential step in such sorting, but the molecular mechanism of this process remains to be elucidated. Here, we show that actin is required for efficient recycling and endosomal maturation by producing a motile force. Perturbation of actin dynamics by drugs induced a few enlarged EEs containing several degradative vacuoles and also interfered with their transporting ability. Actin repolymerization induced by washout of the drug caused the vacuoles to dissociate and individually translocate toward the perinuclear region. We further elucidated that cortactin, an actin-nucleating factor, was required for transporting contents from within EEs. Actin filaments regulated by cortactin may provide a motile force for efficient sorting within early endosomes. These data suggest that actin filaments coordinate with microtubules to mediate segregation in EEs.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Endosomes/metabolism , Microtubules/metabolism , Transport Vesicles/metabolism , Cortactin/antagonists & inhibitors , Cortactin/genetics , Cortactin/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , HeLa Cells , Humans , Protein Transport , RNA, Small Interfering/genetics , Recycling
4.
Mol Cell Biol ; 30(21): 5057-70, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20805359

ABSTRACT

The cortactin oncoprotein is frequently overexpressed in head and neck squamous cell carcinoma (HNSCC), often due to amplification of the encoding gene (CTTN). While cortactin overexpression enhances invasive potential, recent research indicates that it also promotes cell proliferation, but how cortactin regulates the cell cycle machinery is unclear. In this article we report that stable short hairpin RNA-mediated cortactin knockdown in the 11q13-amplified cell line FaDu led to increased expression of the Cip/Kip cyclin-dependent kinase inhibitors (CDKIs) p21(WAF1/Cip1), p27(Kip1), and p57(Kip2) and inhibition of S-phase entry. These effects were associated with increased binding of p21(WAF1/Cip1) and p27(Kip1) to cyclin D1- and E1-containing complexes and decreased retinoblastoma protein phosphorylation. Cortactin regulated expression of p21(WAF1/Cip1) and p27(Kip1) at the transcriptional and posttranscriptional levels, respectively. The direct roles of p21(WAF1/Cip1), p27(Kip1), and p57(Kip2) downstream of cortactin were confirmed by the transient knockdown of each CDKI by specific small interfering RNAs, which led to partial rescue of cell cycle progression. Interestingly, FaDu cells with reduced cortactin levels also exhibited a significant diminution in RhoA expression and activity, together with decreased expression of Skp2, a critical component of the SCF ubiquitin ligase that targets p27(Kip1) and p57(Kip2) for degradation. Transient knockdown of RhoA in FaDu cells decreased expression of Skp2, enhanced the level of Cip/Kip CDKIs, and attenuated S-phase entry. These findings identify a novel mechanism for regulation of proliferation in 11q13-amplified HNSCC cells, in which overexpressed cortactin acts via RhoA to decrease expression of Cip/Kip CDKIs, and highlight Skp2 as a downstream effector for RhoA in this process.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/physiopathology , Cell Cycle/genetics , Cell Cycle/physiology , Cortactin/genetics , Cortactin/physiology , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/physiology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/physiopathology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/physiology , Base Sequence , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human, Pair 11/genetics , Cortactin/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/physiology , DNA Primers/genetics , Gene Amplification , Gene Expression , Gene Knockdown Techniques , Head and Neck Neoplasms/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , RNA, Small Interfering/genetics
5.
J Biol Chem ; 285(39): 29911-24, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20650888

ABSTRACT

The aim of this study is to identify the exact mechanism(s) by which cytoskeletal structures are modulated during bone resorption. In this study, we have shown the possible role of different actin-binding and signaling proteins in the regulation of sealing ring formation. Our analyses have demonstrated a significant increase in cortactin and a corresponding decrease in L-plastin protein levels in osteoclasts subjected to bone resorption for 18 h in the presence of RANKL, M-CSF, and native bone particles. Time-dependent changes in the localization of L-plastin (in actin aggregates) and cortactin (in the sealing ring) suggest that these proteins may be involved in the initial and maturation phases of sealing ring formation, respectively. siRNA to cortactin inhibits this maturation process but not the formation of actin aggregates. Osteoclasts treated as above but with TNF-α demonstrated very similar effects as observed with RANKL. Osteoclasts treated with a neutralizing antibody to TNF-α displayed podosome-like structures in the entire subsurface and at the periphery of osteoclast. It is possible that TNF-α and RANKL-mediated signaling may play a role in the early phase of sealing ring configuration (i.e. either in the disassembly of podosomes or formation of actin aggregates). Furthermore, osteoclasts treated with alendronate or αv reduced the formation of the sealing ring but not actin aggregates. The present study demonstrates a novel mechanistic link between L-plastin and cortactin in sealing ring formation. These results suggest that actin aggregates formed by L-plastin independent of integrin signaling function as a core in assembling signaling molecules (integrin αvß3, Src, cortactin, etc.) involved in the maturation process.


Subject(s)
Bone Resorption/metabolism , Cortactin/metabolism , Cytoskeleton/metabolism , Osteoclasts/metabolism , Phosphoproteins/metabolism , Actins/genetics , Actins/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Bone Resorption/genetics , Cells, Cultured , Cortactin/antagonists & inhibitors , Cortactin/genetics , Cytoskeletal Proteins , Cytoskeleton/genetics , Integrins/genetics , Integrins/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Microfilament Proteins , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , RANK Ligand , RNA, Small Interfering , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/pharmacology
6.
Cancer Res ; 70(12): 5136-46, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501830

ABSTRACT

CUB domain-containing protein 1 (CDCP1) is a membrane protein that is highly expressed in several solid cancers. We reported previously that CDCP1 regulates anoikis resistance as well as cancer cell migration and invasion, although the underlying mechanisms have not been elucidated. In this study, we found that expression of CDCP1 in pancreatic cancer tissue was significantly correlated with overall survival and that CDCP1 expression in pancreatic cancer cell lines was relatively high among solid tumor cell lines. Reduction of CDCP1 expression in these cells suppressed extracellular matrix (ECM) degradation by inhibiting matrix metalloproteinase-9 secretion. Using the Y734F mutant of CDCP1, which lacks the tyrosine phosphorylation site, we showed that CDCP1 regulates cell migration, invasion, and ECM degradation in a tyrosine phosphorylation-dependent manner and that these CDCP1-associated characteristics were inhibited by blocking the association of CDCP1 and protein kinase Cdelta (PKCdelta). CDCP1 modulates the enzymatic activity of PKCdelta through the tyrosine phosphorylation of PKCdelta by recruiting PKCdelta to Src family kinases. Cortactin, which was detected as a CDCP1-dependent binding partner of PKCdelta, played a significant role in migration and invasion but not in ECM degradation of pancreatic cells. These results suggest that CDCP1 expression might play a crucial role in poor outcome of pancreatic cancer through promotion of invasion and metastasis and that molecules blocking the expression, phosphorylation, or the PKCdelta-binding site of CDCP1 are potential therapeutic candidates.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Extracellular Matrix/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Antigens, CD/genetics , Antigens, Neoplasm , Blotting, Western , Cell Adhesion , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Proliferation , Cortactin/antagonists & inhibitors , Cortactin/genetics , Cortactin/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lymphatic Metastasis , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Phosphorylation , Prognosis , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Tyrosine/metabolism
7.
Pathol Oncol Res ; 16(4): 523-31, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20069395

ABSTRACT

Cortactin, an F-actin binding protein, stabilizes F-actin networks and promotes actin polymerization by activating the Arp2/3 complex. Overexpression of cortactin has been reported in several human cancers. Cortactin stimulates cell migration, invasion, and experimental metastasis. However, the underlying mechanism is not still understood. In the present study, we therefore evaluated the possibility that cortactin could be appropriate as a molecular target for cancer gene therapy. In 70 primary oral squamous cell carcinomas and 10 normal oral mucosal specimens, cortactin expression was evaluated by immunological analyses, and the correlations of the overexpression of cortactin with clinicopathologic factors were evaluated. Overexpression of cortactin was detected in 32 of 70 oral squamous cell carcinomas; significantly more frequently than in normal oral mucosa. Cortactin overexpression was more frequent in higher grade cancers according to T classification, N classifications, and invasive pattern. Moreover, RNAi-mediated decrease in cortactin expression reduced invasion. Downregulation of cortactin expression increased the expression levels of E-cadherin, ß-catenin, and EpCAM. The siRNA of cortactin also reduced PTHrP expression via EGF signaling. These results consistently indicate that the overexpression of cortactin is strongly associated with an aggressive phenotype of oral squamous cell carcinoma. In conclusion, we propose that cortactin could be a potential molecular target of gene therapy by RNAi targeting in oral squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cortactin/biosynthesis , Mouth Neoplasms/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cortactin/antagonists & inhibitors , Cortactin/genetics , Cortactin/metabolism , ErbB Receptors/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Staging , Parathyroid Hormone-Related Protein/biosynthesis , Prognosis , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transfection
8.
J Cell Sci ; 121(Pt 3): 369-78, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18198194

ABSTRACT

Invadopodia are proteolytically active protrusions formed by invasive tumoral cells when grown on an extracellular matrix (ECM) substratum. Although many molecular components have been defined, less is known of the formation and regulation of invadopodia. The multidomain protein cortactin, which is involved in the regulation of actin polymerisation, is one such component, but how cortactin is modulated to control the formation of invadopodia has not been elucidated. Here, a new invadopodia synchronization protocol is used to show that the cortactin N-terminal acidic and SH3 domains, involved in Arp2/3 complex and N-WASP binding and activation, respectively, are both required for invadopodia biogenesis. In addition, through a combination of RNA interference and a wide array of cortactin phosphorylation mutants, we were able to show that three convergent regulatory inputs based on the regulation of cortactin phosphorylation by Src-family kinases, Erk1/Erk2 and PAK are necessary for invadopodia formation and extracellular matrix degradation. These findings suggest that cortactin is a scaffold protein bringing together the different components necessary for the formation of the invadopodia, and that a fine balance between different phosphorylation events induces subtle changes in structure to calibrate cortactin function.


Subject(s)
Cell Surface Extensions/physiology , Cortactin/physiology , Extracellular Matrix/physiology , Neoplasm Invasiveness/physiopathology , Actin-Related Protein 2-3 Complex/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Surface Extensions/pathology , Cortactin/antagonists & inhibitors , Cortactin/chemistry , Cortactin/genetics , DNA Primers/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Melanoma/pathology , Melanoma/physiopathology , Protein Structure, Tertiary , RNA Interference , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , p21-Activated Kinases/metabolism , src Homology Domains , src-Family Kinases/metabolism
9.
J Immunol ; 177(9): 6440-9, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17056576

ABSTRACT

Endothelial cell ICAM-1 interacts with leukocyte beta(2) integrins to mediate adhesion and transmit outside-in signals that facilitate leukocyte transmigration. ICAM-1 redistribution and clustering appear necessary for leukocyte transmigration, but the mechanisms controlling ICAM-1 redistribution and clustering have not been identified. We recently reported that Src kinase phosphorylation of endothelial cortactin regulates polymorphonuclear cell (PMN) transmigration. In this study, we tested the hypotheses that the Src family kinase-cortactin pathway mediates association of ICAM-1 with the actin cytoskeleton and that this association is required for ICAM-1 clustering and leukocyte transmigration. Cross-linking ICAM-1 induced cytoskeletal remodeling and a decrease in ICAM-1 lateral mobility, as assessed by fluorescence recovery after photobleaching. Cytoskeletal remodeling after ICAM-1 cross-linking was reduced by knockdown of cortactin by small interfering RNA, by expression of a cortactin mutant deficient in Src phosphorylation sites (cortactin3F), and by the Src kinase inhibitor PP2. Pretreatment of cytokine-activated human endothelial monolayers with cortactin small interfering RNA significantly decreased both actin and ICAM-1 clustering around adherent PMN and the formation of actin-ICAM-1 clusters required for PMN transmigration. Our data suggest a model in which tyrosine phosphorylation of cortactin dynamically links ICAM-1 to the actin cytoskeleton, enabling ICAM-1 to form clusters and facilitate leukocyte transmigration.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Adhesion , Cell Movement , Cortactin/metabolism , Intercellular Adhesion Molecule-1/metabolism , Neutrophils/immunology , Actins/metabolism , Cell Adhesion/genetics , Cell Movement/genetics , Cells, Cultured , Cortactin/antagonists & inhibitors , Cortactin/genetics , Cytoskeleton/metabolism , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Humans , Phosphorylation , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Tyrosine , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...