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.
Cancer Res ; 61(19): 7079-90, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585739

ABSTRACT

Elevated focal adhesion kinase (FAK) expression in human tumor cells has been correlated with an increased cell invasion potential. In cell culture, studies with FAK-null fibroblasts have shown that FAK function is required for cell migration. To determine the role of elevated FAK expression in facilitating epidermal growth factor (EGF)-stimulated human adenocarcinoma (A549) cell motility, antisense oligonucleotides were used to reduce FAK protein expression >75%. Treatment of A549 cells with FAK antisense (ISIS 15421) but not a mismatched control (ISIS 17636) oligonucleotide resulted in reduced EGF-stimulated p130(Cas)-Src complex formation, c-Jun NH(2)-terminal kinase (JNK) activation, directed cell motility, and serum-stimulated cell invasion through Matrigel. Because residual FAK protein in ISIS 15421-treated A549 cells was highly phosphorylated at the Tyr-397/Src homology (SH)2 binding site, expression of the FAK COOH-terminal domain (FRNK) was also used as an inhibitor of FAK function. Adenoviral-mediated infection and expression of FRNK promoted FAK dephosphorylation at Tyr-397, resulted in reduced EGF-stimulated JNK as well as extracellular-regulated kinase 2 (ERK2) kinase activation, inhibited matrix metalloproteinase-9 (MMP-9) secretion, and potently blocked both random and EGF-stimulated A549 cell motility. Equivalent expression of a FRNK (S-1034) point-mutant that did not promote FAK dephosphorylation also did not affect EGF-stimulated signaling or cell motility. Dose-dependent reduction in EGF-stimulated A549 motility was observed with the PD98059 MEK1 inhibitor and the batimastat (BB-94) inhibitor of MMP activity, but not with the SB203580 inhibitor of p38 kinase. Finally, comparisons between normal, FAK-null, and FAK-reconstituted fibroblasts revealed that FAK enhanced EGF-stimulated JNK and ERK2 kinase activation that was required for cell motility. These data indicate that FAK functions as an important signaling platform to coordinate EGF-stimulated cell migration in human tumor cells and support a role for inhibitors of FAK expression or activity in the control of neoplastic cell invasion.


Subject(s)
Adenocarcinoma/enzymology , Cell Movement/physiology , Epidermal Growth Factor/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Adenocarcinoma/pathology , Cell Movement/drug effects , Enzyme Activation , Epidermal Growth Factor/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Tumor Cells, Cultured
2.
J Cell Sci ; 113 ( Pt 20): 3673-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017882

ABSTRACT

Focal adhesion kinase (FAK) is activated and localized at focal adhesions upon cell adhesion to extracellular matrices. Cells lacking FAK show increased focal adhesion number and decreased cell migration, functions that are regulated by the small GTPase Rho. We now report that fibroblasts from FAK-/- mice failed to transiently inhibit Rho activity when plated on fibronectin. Re-expression of FAK restored normal Rho regulation. Turnover of focal adhesions correlated inversely with Rho activity. The presence or absence of FAK was mimicked by inhibiting or activating Rho, respectively. These data suggest that loss of FAK resulting in constitutive activation of Rho and inhibition of focal adhesion turnover can account for deficiencies in cell migration and embryonic lethality of the FAK knockout.


Subject(s)
Focal Adhesions , Protein-Tyrosine Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Movement , Cell Size , Cells, Cultured , Cytoskeleton/ultrastructure , Enzyme Activation , Fibroblasts , Fibronectins , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , rho GTP-Binding Proteins/antagonists & inhibitors
3.
Nat Cell Biol ; 2(5): 249-56, 2000 May.
Article in English | MEDLINE | ID: mdl-10806474

ABSTRACT

Here we show that cells lacking focal adhesion kinase (FAK) are refractory to motility signals from platelet-derived and epidermal growth factors (PDGF and EGF respectively), and that stable re-expression of FAK rescues these defects. FAK associates with activated PDGF- and EGF-receptor (PDGFR and EGFR) signalling complexes, and expression of the band-4.1-like domain at the FAK amino terminus is sufficient to mediate an interaction with activated EGFR. However, efficient EGF-stimulated cell migration also requires FAK to be targeted, by its carboxy-terminal domain, to sites of integrin-receptor clustering. Although the kinase activity of FAK is not needed to promote PDGF- or EGF-stimulated cell motility, kinase-inactive FAK is transphosphorylated at the indispensable Src-kinase-binding site, FAK Y397, after EGF stimulation of cells. Our results establish that FAK is an important receptor-proximal link between growth-factor-receptor and integrin signalling pathways.


Subject(s)
Cell Movement/physiology , Integrins/metabolism , MAP Kinase Signaling System/physiology , Platelet-Derived Growth Factor/pharmacology , Protein-Tyrosine Kinases/metabolism , Cell Movement/drug effects , Cells, Cultured , Epidermal Growth Factor/pharmacology , ErbB Receptors/physiology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Humans , MAP Kinase Signaling System/drug effects , Mutagenesis/physiology , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Receptors, Platelet-Derived Growth Factor/physiology , src-Family Kinases/metabolism
4.
J Cell Sci ; 112 ( Pt 16): 2677-91, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10413676

ABSTRACT

FAK localizes to sites of transmembrane integrin receptor clustering and facilitates intracellular signaling events. FAK-null (FAK-) fibroblasts exhibit a rounded morphology, defects in cell migration, and an elevated number of cell-substratum contact sites. Here we show that stable re-expression of epitope-tagged FAK reversed the morphological defects of the FAK- cells through the dynamic regulation of actin structures and focal contact sites in fibronectin (FN) stimulated cells. FAK re-expressing fibroblasts (clones DA2 and DP3) exhibit a characteristic fibrillar shape and display indistinguishable FN receptor-stimulated migration properties compared to normal fibroblasts. Expression of various FAK mutants in the FAK- cells showed that FAK kinase activity, the Tyr-397/SH2 domain binding site, and the first proline-rich SH3 binding region in the FAK C-terminal domain were individually needed to promote full FAK-mediated FAK- cell migration to FN whereas direct paxillin binding to FAK was not required. Expression of the FAK Phe-397 mutant did not promote FAK- cell migration and overexpression of p50(csk) in DA2 cells inhibited migration to FN suggesting that Src-family PTKs play important roles in FAK-mediated motility events. Expression of the FAK C-terminal domain, FRNK, promoted FAK dephosphorylation at Tyr-397 and potently blocked FAK-mediated cell migration. This dominant-negative effect of FRNK was reversed by a point mutation (Leu-1034 to Ser) which prevented FRNK localization to focal contact sites. Our results show that FAK functions as a key regulator of fibronectin receptor stimulated cell migration events through the recruitment of both SH2 and SH3 domain-containing signaling proteins to sites of integrin receptor clustering.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Integrins/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Actins/analysis , Actins/metabolism , Animals , Cell Adhesion Molecules/analysis , Cell Movement/drug effects , Cells, Cultured , Cytoskeletal Proteins/analysis , Epitopes/genetics , Fibroblasts/chemistry , Fibroblasts/cytology , Fibroblasts/enzymology , Fibronectins/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Gene Expression Regulation, Enzymologic , Mice , Mice, Knockout , Mutagenesis, Site-Directed/physiology , Paxillin , Phenotype , Phosphoproteins/analysis , Phosphorylation , Proline/metabolism , Protein Binding/physiology , Protein-Tyrosine Kinases/analysis , Talin/analysis , Transfection , Vinculin/analysis , src Homology Domains/physiology
5.
Prog Biophys Mol Biol ; 71(3-4): 435-78, 1999.
Article in English | MEDLINE | ID: mdl-10354709

ABSTRACT

Integrin receptor binding to extracellular matrix proteins generates intracellular signals via enhanced tyrosine phosphorylation events that are important for cell growth, survival, and migration. This review will focus on the functions of the focal adhesion kinase (FAK) protein-tyrosine kinase (PTK) and its role in linking integrin receptors to intracellular signaling pathways. FAK associates with several different signaling proteins such as Src-family PTKs, p130Cas, Shc, Grb2, PI 3-kinase, and paxillin. This enables FAK to function within a network of integrin-stimulated signaling pathways leading to the activation of targets such as the ERK and JNK/mitogen-activated protein kinase pathways. Focus will be placed on the structural domains and sites of FAK tyrosine phosphorylation important for FAK-mediated signaling events and how these sites are conserved in the FAK-related PTK, Pyk2. We will review what is known about FAK activation by integrin receptor-mediated events and also non-integrin stimuli. In addition, we discuss the emergence of a consensus FAK substrate phosphorylation sequence. Emphasis will also be placed on the role of FAK in generating cell survival signals and the cleavage of FAK during caspase-mediated apoptosis. An in-depth discussion will be presented of integrin-stimulated signaling events occurring in the FAK knockout fibroblasts (FAK-) and how these cells exhibit deficits in cell migration. FAK re-expression in the FAK- cells confirms the role of this PTK in the regulation of cell morphology and in promoting cell migration events. In addition, these results reinforce the potential role for FAK in promoting an invasive phenotype in human tumors.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Cell Adhesion Molecules/chemistry , Extracellular Matrix Proteins/physiology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Integrins/physiology , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein-Tyrosine Kinases/chemistry
6.
EMBO J ; 17(20): 5933-47, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9774338

ABSTRACT

The focal adhesion kinase (FAK) protein-tyrosine kinase (PTK) links transmembrane integrin receptors to intracellular signaling pathways. We show that expression of the FAK-related PTK, Pyk2, is elevated in fibroblasts isolated from murine fak-/- embryos (FAK-) compared with cells from fak+/+ embryos (FAK+). Pyk2 was localized to perinuclear regions in both FAK+ and FAK- cells. Pyk2 tyrosine phosphorylation was enhanced by fibronectin (FN) stimulation of FAK- but not FAK+ cells. Increased Pyk2 tyrosine phosphorylation paralleled the time-course of Grb2 binding to Shc and activation of ERK2 in FAK- cells. Pyk2 in vitro autophosphorylation activity was not enhanced by FN plating of FAK- cells. However, Pyk2 associated with active Src-family PTKs after FN but not poly-L-lysine replating of the FAK- cells. Overexpression of both wild-type (WT) and kinase-inactive (Ala457), but not the autophosphorylation site mutant (Phe402) Pyk2, enhanced endogenous FN-stimulated c-Src in vitro kinase activity in FAK- cells, but only WT Pyk2 overexpression enhanced FN-stimulated activation of co-transfected ERK2. Interestingly, Pyk2 overexpression only weakly augmented FAK- cell migration to FN whereas transient FAK expression promoted FAK- cell migration to FN efficiently compared with FAK+ cells. Significantly, repression of endogenous Src-family PTK activity by p50(csk) overexpression inhibited FN-stimulated cell spreading, Pyk2 tyrosine phosphorylation, Grb2 binding to Shc, and ERK2 activation in the FAK- but not in FAK+ cells. These studies show that Pyk2 and Src-family PTKs combine to promote FN-stimulated signaling events to ERK2 in the absence of FAK, but that these signaling events are not sufficient to overcome the FAK- cell migration defects.


Subject(s)
Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/physiology , Cell Movement/physiology , Fibronectins/metabolism , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , src-Family Kinases/physiology , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cells, Cultured , Fibroblasts , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Mice , Mitogen-Activated Protein Kinase 1 , Molecular Sequence Data , Protein-Tyrosine Kinases/biosynthesis , Rabbits , Vinculin/analysis
7.
Toxicol Appl Pharmacol ; 142(1): 106-15, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9007039

ABSTRACT

Hepatocytes exposed to the cytokines tumor necrosis factor alpha (TNFalpha) and interferon gamma (IFNgamma), in combination with a nontoxic dose of lead (Pb), exhibited significantly greater cytotoxicity compared to cytokine-treated hepatocytes after 24 hr treatment. Concentrations of Pb which interact with the cytokines to cause cytotoxicity stimulated hepatocytes cultured at this high cell density (63,830 cells/cm2) to undergo active DNA synthesis. DNA synthesis was also observed in hepatocytes plated at a low cell densities (21,276 cells/cm2) and these cells were sensitive to the toxic effects of the cytokines in the absence of Pb. Under conditions of active DNA synthesis, induced by either Pb or cell density, cytokine-induced toxicity appeared to be the result of extensive DNA fragmentation. Both DNA fragmentation and cytotoxicity were inhibited by treatment with an antioxidant mixture. Pb potentiated cytokine-induced oxidative stress within hepatocytes as indicated by decreased intracellular glutathione (GSH) and increased efflux of oxidized glutathione (GSSG) prior to cytotoxicity. The combination of cytokines and Pb also caused a significant decline in intracellular ATP concentrations prior to the onset of cytotoxicity. ATP concentrations were not altered by treatment with Pb or cytokines alone. Pb treatment did not alter total protein synthesis within hepatocytes. These results indicate that the cytotoxic interaction between Pb and the cytokines TNFalpha and IFNgamma may be mediated by oxidative DNA damage resulting from cytokine-induced oxidative stress and stimulation of mitogenic signals.


Subject(s)
DNA Damage , Interferon-gamma/toxicity , Liver/drug effects , Organometallic Compounds/toxicity , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/toxicity , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Cells, Cultured , DNA Fragmentation/drug effects , DNA Repair/drug effects , DNA Replication/drug effects , Drug Synergism , Glutathione/metabolism , Interferon-gamma/pharmacology , L-Lactate Dehydrogenase/analysis , Liver/cytology , Male , Mice , Organometallic Compounds/pharmacology , Protein Biosynthesis , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
8.
Arch Biochem Biophys ; 316(2): 699-706, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7532384

ABSTRACT

Regulation of induced nitric oxide synthase in rat hepatocyte primary cultures was explored. Nitric oxide synthase (NOS) induction by tumor necrosis factor-alpha (TNF alpha) is synergized by interferon-gamma, and both NOS activity and gene expression are maximal by 10 h and maintained through 24 h. Glutathione depletion by diethylmaleate, which conjugates reduced glutathione, 1,3-bis(chloroethyl)-1-nitrosourea (BCNU), a glutathione reductase inhibitor, or buthionine sulfoxamine, a glutathione synthesis inhibitor, abolishes or reduces NOS induction in TNF alpha-treated hepatocytes, whereas N-acetylcysteine has little effect. Thus, reduced glutathione is critical to NOS mRNA induction and activity in TNF alpha-treated hepatocytes. NOS induction in TNF alpha-treated cells is reduced by rotenone, a mitochondrial complex 1 inhibitor. Concurrent treatment with TNF alpha and the antioxidant, Trolox, or the iron-chelating agent, desferrioxamine, also reduces NOS activity. Dithiothreitol, a thiol antioxidant, reduced TNF alpha induction of NOS. Trolox and BCNU, combined, blocked TNF alpha stimulation of NOS greater than either agent alone. These results suggest that TNF alpha increases mitochondrial production of reactive oxygen intermediates (ROI), which contributes to NOS induction. Hepatocytes exposed to extracellular ROI generation through a xanthine/xanthine oxidase superoxide-generating system expressed increased NOS activity and mRNA levels. NOS induction by superoxide also requires reduced glutathione since diethylmaleate blocks induction by xanthine/xanthine oxidase while N-acetylcysteine elevates NOS expression. Thus, the generation of ROI by cytokines or other physiological processes stimulates the induction of NOS and this process is regulated by cellular levels of reduced glutathione.


Subject(s)
Amino Acid Oxidoreductases/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Liver/enzymology , Amino Acid Oxidoreductases/genetics , Animals , Carmustine/pharmacology , Glutathione/pharmacology , Interferon-gamma/pharmacology , Liver/cytology , Liver/drug effects , Maleates/pharmacology , Nitric Oxide Synthase , Oxidation-Reduction , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/pharmacology , Superoxides/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Xanthine , Xanthine Oxidase/pharmacology , Xanthines/pharmacology
9.
J Invest Dermatol ; 97(6): 1019-23, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1660906

ABSTRACT

The relationship between ultraviolet irradiation, interleukin-1 production, and inflammatory sequelae and the pharmacologic inhibition of these events was investigated in Balb/c mice exposed to ultraviolet irradiation from a bank of six Westinghouse FS40 sunlamps. The resulting edema (66% increase), inflammatory cell infiltration, and rise in the acute-phase reactant (fourfold) serum amyloid P component was preceded by the activation of the interleukin-1 beta gene and enhanced product formation. Administration of dexamethasone, which is known to inhibit interleukin-1 production, inhibited the inflammatory response to ultraviolet irradiation. Thus, production of interleukin-1 may be one of the initial events leading to the consequences of ultraviolet irradiation exposure.


Subject(s)
Interleukin-1/genetics , Skin/radiation effects , Ultraviolet Rays , Animals , Blotting, Northern , Dermatitis/etiology , Dexamethasone/pharmacology , Gene Expression Regulation/radiation effects , Interleukin-1/metabolism , Male , Mice , Mice, Inbred BALB C , Peroxidase/analysis , Serum Amyloid P-Component/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...