Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Allergy ; 68(4): 481-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23409786

ABSTRACT

BACKGROUND: One of the most promising strategies to face the increasing asthma prevalence and to prevent disease development might be an early contact with microbial compounds. However, little is known about an interaction between an early-life contact to microbial compounds leading to asthma protection in the offspring and a co-exposure to allergy-promoting pollutants. METHODS: Pregnant BALB/c mice were repeatedly exposed to aerosolized endotoxin (lipopolysaccharide, LPS). The offspring was further exposed to aerosolized LPS before allergen sensitization with ovalbumin (OVA). Some of the mice were co-exposed to mycotoxins or diesel exhaust particles (DEP) during pregnancy. The 6-week-old offspring was immunized with OVA and analyzed in a murine asthma model. RESULTS: While the offspring of naïve mothers developed an asthma-like phenotype, the offspring of mice perinatally exposed to LPS was significantly protected. Co-exposure of mice to mycotoxins or DEP during pregnancy inhibited the LPS-induced protection leading to the development of eosinophilic airway inflammation, airway hyperactivity, and increased antigen-specific IgE levels in the offspring. Furthermore, the asthma-preventive effect of perinatal LPS exposure was IFN-gamma dependent. Additionally, the IFN-gamma promoter of CD4+ T cells in the LPS-exposed offspring revealed a significant protection against loss of histone 4 acetylation, which was abolished after prenatal co-exposure to pollutants. Prenatal treatment of mice with the antioxidant N-acetylcysteine reversed the pollutant-induced increased asthma risk in the offspring. CONCLUSION: Our results show that exposure to pollutants during pregnancy may cause the development of allergic asthma in the offspring by inhibiting the endotoxin-induced perinatal asthma protection.


Subject(s)
Air Pollutants/immunology , Asthma/immunology , Asthma/prevention & control , Acetylation , Acetylcysteine/pharmacology , Allergens/immunology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Antioxidants/pharmacology , Asthma/genetics , Disease Models, Animal , Endotoxins/adverse effects , Endotoxins/immunology , Epigenesis, Genetic , Female , Histones/metabolism , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/genetics , Lipopolysaccharides/immunology , Maternal Exposure , Mice , Mycotoxins/adverse effects , Mycotoxins/immunology , Ovalbumin/immunology , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Promoter Regions, Genetic
2.
Clin Exp Allergy ; 38(12): 1950-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18778271

ABSTRACT

BACKGROUND: Maternal, more than paternal, asthma is a risk factor for the development of asthma in children. Recently, epidemiologic studies have shown that environmental exposures during pregnancy might influence the development of childhood asthma and allergies. OBJECTIVE: The aim of the present study was to investigate whether the induction of tolerance against a specific antigen during pregnancy prevents in the offspring the development of allergic asthma in response to this antigen. METHODS: Balb/c mice were orally tolerized with ovalbumin (OVA) during pregnancy. The offspring of tolerized and naïve mothers were immunized with OVA at 6 weeks and 4 months of age and analysed in our murine asthma model. RESULTS: While the offspring of naïve mice developed increased AHR, eosinophilic airway inflammation, T-helper type 2 cytokine production and high serum IgE levels in response to OVA sensitization, the offspring of tolerized mice were almost completely protected from asthma, even when immunized as late as 4 months after birth. Breastfeeding was crucial for protection because tolerance was only observed when the offspring were nursed by their own mothers and not when nursed by naïve wet-nurses. Allergen-specific IgG(1) antibodies were exclusively increased in the breast milk of tolerant mothers and serum of protected pups, indirectly supporting its important role in tolerance transfer from the mother to the offspring. Sensitization of the F1 generation from OVA-tolerized mothers with a heterologous allergen enhanced the immune response to this antigen. CONCLUSION: Our results demonstrate that mucosal allergen contact during pregnancy modifies the asthma and allergy risk of the offspring mediated via breast milk. This observation may suggest that the time window for primary prevention strategies starts even before early childhood during pregnancy.


Subject(s)
Asthma/immunology , Immune Tolerance , Immunity, Maternally-Acquired , Immunization , Milk, Human/immunology , Allergens/adverse effects , Animals , Animals, Suckling , Antibodies/blood , Antibodies/immunology , Antibody Specificity , Antigens/administration & dosage , Antigens/immunology , Asthma/etiology , Disease Models, Animal , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/adverse effects , Ovalbumin/immunology , Pregnancy
3.
Clin Exp Allergy ; 37(9): 1374-85, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17845419

ABSTRACT

BACKGROUND: Allergic asthma is a T-helper type 2 (Th2) cell-mediated chronic disease that is characterized by airway hyperreactivity (AHR) and chronic eosinophilic airway inflammation. Several studies suggest co-stimulatory molecules like CD137 as potential targets for therapeutic interventions in allergic airway disease. Recently, we could show in a murine asthma model that administration of an agonistic antibody against the receptor of the co-stimulatory molecule CD137 prevented and even reversed an already-established asthma phenotype. OBJECTIVE: The purpose of this study was to analyse the effect of stimulation of the CD137 ligand by a monoclonal antibody (CD137L mAb). METHODS: To induce an asthma-like phenotype, BALB/c mice were sensitized to ovalbumin (OVA), followed by an intrapulmonary allergen challenge. Anti-CD137L or control mAb were applied 1 day before OVA immunization or after the asthma phenotype was already established. RESULTS: Stimulation of the CD137L instead of the receptor by CD137L mAb prevents the development of an asthma-like phenotype but does not reverse established disease. While the receptor-mediated effect is partly mediated by anergy of CD4(+) T cells and partly by induction of IFN-gamma-producing CD8(+) T cells, the effect of the CD137L mAb is completely dependent on IFN-gamma-producing CD8(+) T cells: blockade of IFN-gamma and depletion of CD8(+) T cells fully abrogated the observed protective effect. In vitro experiments showed that the anti-CD137L mAb ligates directly to CD8(+) T cells and induces the generation of IFN-gamma by this cell population. CONCLUSION: Our results demonstrate that anti-CD137L mAb prevents disease development via IFN-gamma-producing CD8(+) T cells but is inferior to stimulation of the receptor that reverses established disease by a mechanism including CD4(+) T cell anergy.


Subject(s)
4-1BB Ligand/immunology , Antibodies, Monoclonal/administration & dosage , Asthma/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/biosynthesis , Animals , Asthma/prevention & control , Disease Models, Animal , Female , Immunity, Cellular , Mice , Mice, Inbred BALB C , Th2 Cells/immunology
4.
Clin Exp Allergy ; 35(8): 1003-13, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16120081

ABSTRACT

BACKGROUND: Allergen-induced T-helper type 2 (Th2) responses can be inhibited with Th1 directing vaccines. However, studies comparing the efficacy of the different adjuvants have not been performed in detail. OBJECTIVE: For this reason we compare the effects of live Bacillus-Calmette-Guerin(BCG), heat-killed (hk)-BCG, CpG-ODN (oligodeoxynucleotide) or PPD on the development of allergen-induced Th2 responses in mice. METHODS: Ovalbumin (OVA)-specific allergic responses were induced in C57BL/6 mice by two intraperitoneally (i.p.) applications of OVA/alum followed by the intranasal challenge with OVA. The different Th1-inducing adjuvants were applied to the mice together with OVA/alum i.p. during the OVA-sensitization period and, subsequently, different parameters of allergic immune responses were evaluated. RESULTS: All the adjuvants were effective in inhibiting the development of allergen-induced airway eosinophilia, mucous production and, with the exception of PPD, also airway hyper-reactivity, when they were applied together with OVA/alum. However, allergen-specific IgG1 and IgE serum levels were only reduced in live BCG- and PPD-treated mice. Suppression of airway eosinophilia was not observed in IFN-gamma- or IL-12-deficient mice (hk-BCG, CpG-ODN and PPD). Interestingly, live BCG was still able to suppress allergen-induced Th2 responses in the absence of either IFN-gamma or IL-12. When mice vaccinated with the different adjuvants together with OVA/alum were subjected to a second period of OVA/alum immunization, only live and hk-BCG were able to efficiently suppress the development of airway inflammation. This effect could be adoptively transferred by splenic CD4(+) T cells. CONCLUSIONS: Taken together our data suggest that live BCG>hk-BCG>CpG-ODN >PPD are effective in suppressing allergen-induced Th2 responses. The degree of suppression and the component of the Th2 response affected (airway inflammation vs. the production of allergen-specific IgE and IgG1) were dependent upon the adjuvant used and how it was applied. Our results contribute to the design of novel vaccines protecting humans from developing allergic disorders.


Subject(s)
Allergens/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Vaccination/methods , Adjuvants, Immunologic , Adoptive Transfer/methods , Animals , BCG Vaccine/immunology , Cells, Cultured , Eosinophils/immunology , Female , Immune Tolerance/immunology , Immunoglobulin E/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Oligodeoxyribonucleotides/immunology , Ovalbumin/immunology , Respiratory System/immunology , Tuberculin/immunology
5.
Mol Cell Biol ; 21(22): 7641-52, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11604500

ABSTRACT

Tyrosine phosphorylation of CAS (Crk-associated substrate, p130(Cas)) has been implicated as a key signaling step in integrin control of normal cellular behaviors, including motility, proliferation, and survival. Aberrant CAS tyrosine phosphorylation may contribute to cell transformation by certain oncoproteins, including v-Crk and v-Src, and to tumor growth and metastasis. The CAS substrate domain (SD) contains 15 Tyr-X-X-Pro motifs, which are thought to represent the major tyrosine phosphorylation sites and to function by recruiting downstream signaling effectors, including c-Crk and Nck. CAS makes multiple interactions, direct and indirect, with the tyrosine kinases Src and focal adhesion kinase (FAK), and as a result of this complexity, several plausible models have been proposed for the mechanism of CAS-SD phosphorylation. The objective of this study was to provide experimental tests of these models in order to determine the most likely mechanism(s) of CAS-SD tyrosine phosphorylation by FAK and Src. In vitro kinase assays indicated that FAK has a very poor capacity to phosphorylate CAS-SD, relative to Src. However, FAK expression along with Src was found to be important for achieving high levels of CAS tyrosine phosphorylation in COS-7 cells, as well as recovery of CAS-associated Src activity toward the SD. Structure-functional studies for both FAK and CAS further indicated that FAK plays a major role in regulating CAS-SD phosphorylation by acting as a docking or scaffolding protein to recruit Src to phosphorylate CAS, while a secondary FAK-independent mechanism involves Src directly bound to the CAS Src-binding domain (SBD). Our results do not support models in which FAK either phosphorylates CAS-SD directly or phosphorylates CAS-SBD to promote Src binding to this site.


Subject(s)
Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins , Tyrosine/metabolism , src-Family Kinases/metabolism , Amino Acid Motifs , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Crk-Associated Substrate Protein , Focal Adhesion Protein-Tyrosine Kinases , Phosphorylation , Retinoblastoma-Like Protein p130 , src Homology Domains
6.
Proc Natl Acad Sci U S A ; 98(14): 7765-70, 2001 Jul 03.
Article in English | MEDLINE | ID: mdl-11438729

ABSTRACT

Alternative models of cell mechanics depict the living cell as a simple mechanical continuum, porous filament gel, tensed cortical membrane, or tensegrity network that maintains a stabilizing prestress through incorporation of discrete structural elements that bear compression. Real-time microscopic analysis of cells containing GFP-labeled microtubules and associated mitochondria revealed that living cells behave like discrete structures composed of an interconnected network of actin microfilaments and microtubules when mechanical stresses are applied to cell surface integrin receptors. Quantitation of cell tractional forces and cellular prestress by using traction force microscopy confirmed that microtubules bear compression and are responsible for a significant portion of the cytoskeletal prestress that determines cell shape stability under conditions in which myosin light chain phosphorylation and intracellular calcium remained unchanged. Quantitative measurements of both static and dynamic mechanical behaviors in cells also were consistent with specific a priori predictions of the tensegrity model. These findings suggest that tensegrity represents a unified model of cell mechanics that may help to explain how mechanical behaviors emerge through collective interactions among different cytoskeletal filaments and extracellular adhesions in living cells.


Subject(s)
Cell Physiological Phenomena , Cytoskeleton/physiology , Animals , Biomechanical Phenomena , Cytoskeleton/ultrastructure , Green Fluorescent Proteins , Humans , Luminescent Proteins , Models, Biological , Molecular Motor Proteins/physiology
7.
Arterioscler Thromb Vasc Biol ; 20(5): 1209-15, 2000 May.
Article in English | MEDLINE | ID: mdl-10807735

ABSTRACT

Inducible heme oxygenase (HO-1) has recently been recognized as an antioxidant and cytoprotective gene. By use of Western blotting, cell viability analysis, and antisense technique, the present study investigates the involvement of HO-1 in endothelial protection induced by the clinically used nitric oxide (NO) donor molsidomine (specifically, its active metabolite 3-morpholinosydnonimine [SIN-1]) and the second messenger cGMP. In bovine pulmonary artery endothelial cells, SIN-1 and S-nitroso-N-acetyl-D,L-penicillamine (SNAP) at 1 to 100 micromol/L induced the synthesis of HO-1 protein in a concentration-dependent fashion up to 3-fold over basal levels. HO-1 induction by SIN-1 was inhibited in the presence of the NO scavenger phenyl-4,4,5,5,-tetramethylimidazoline-1-oxyl-3-oxide and the soluble guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazole[4, 3-a]quinoxalin-1-one. 8-Bromo-cGMP (1 to 100 micromol/L) and dibutyryl cGMP (1 to 100 micromol/L) as well as the activator of particulate guanylyl cyclase atrial natriuretic peptide (1 to 100 nmol/L) produced increases in HO-1 protein similar to those produced by SIN-1. SIN-1 and 8-bromo-cGMP increased heme oxygenase activity (bilirubin formation). Cytoprotection by NO donors was abrogated in the presence of the heme oxygenase inhibitor tin protoporphyrin IX. Pretreatment of cells with a phosphorothioate-linked HO-1 antisense oligonucleotide prevented protection by SIN-1 or 8-bromo-cGMP against tumor necrosis factor-alpha cytotoxicity, whereas sense and scrambled HO-1 were without effect under these conditions. Our results show for the first time that HO-1 is a cGMP-sensitive endothelial gene and establish conclusively a causal relationship between HO-1 induction and endothelial protection by the NO/cGMP system. By targeting cytoprotective HO-1, NO donors may therefore be expected to induce antioxidant, antiatherogenic, and anti-inflammatory effects.


Subject(s)
Cyclic GMP/pharmacology , Endothelium, Vascular/enzymology , Heme Oxygenase (Decyclizing)/physiology , Nitric Oxide/pharmacology , Animals , Cattle , Cell Survival , Cells, Cultured , Cyclic GMP/analogs & derivatives , Enzyme Induction/drug effects , Heme Oxygenase (Decyclizing)/biosynthesis , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Pulmonary Artery , Second Messenger Systems
8.
Biochem Biophys Res Commun ; 251(2): 460-5, 1998 Oct 20.
Article in English | MEDLINE | ID: mdl-9792796

ABSTRACT

Incubation with TNF-alpha (50 ng/ml) for 72 hours markedly reduced viability of endothelial cells. A 6-hour preincubation with S-nitroso-N-acetyl-D,L-penicillamine (SNAP, 3-100 microM) protected cells in a concentration-dependent manner and decreased TNF-alpha-mediated toxicity by up to 70%. Cytoprotection by SNAP was completely abolished by the adenylyl cyclase inhibitor 2', 5'-dideoxyadenosine and mimicked by 8-bromo cyclic AMP or forskolin. SNAP produced significant increases in cyclic GMP and cyclic AMP, both being abrogated in the presence of the NO scavenger 2-phenyl-4, 4,5,5,-tetramethylimidazoline-1-oxyl-3-oxide (PTIO). Moreover, no endothelial protection by SNAP was detected in the presence of the protein kinase A inhibitor KT5720, whereas the protein kinase G inhibitor KT5823 left cytoprotection virtually unaltered. Our results demonstrate a crucial role for cyclic AMP in mediating NO-induced endothelial protection against TNF-alpha, possibly through cyclic GMP-dependent inhibition of cyclic AMP breakdown. NO-dependent endothelial protection may ultimately result from cyclic AMP-induced up-regulation of antioxidant proteins or down-regulation of cytotoxic processes.


Subject(s)
Carbazoles , Cell Survival/physiology , Cyclic AMP/metabolism , Endothelium, Vascular/physiology , Nitric Oxide/physiology , Penicillamine/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenylyl Cyclase Inhibitors , Alkaloids/pharmacology , Animals , Cattle , Cell Line , Cell Survival/drug effects , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP/metabolism , Cyclic N-Oxides/pharmacology , Dideoxyadenosine/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Kinetics , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacology , Oxadiazoles/pharmacology , Penicillamine/pharmacology , Pulmonary Artery , Pyrroles/pharmacology , Quinoxalines/pharmacology , S-Nitroso-N-Acetylpenicillamine , Tumor Necrosis Factor-alpha/toxicity
9.
Circ Res ; 82(9): 1016-20, 1998 May 18.
Article in English | MEDLINE | ID: mdl-9598599

ABSTRACT

Aspirin has recently been shown to increase endothelial resistance to oxidative damage. However, the mechanism underlying aspirin-induced cytoprotection is still unknown. Using cultured cells, the present study investigates the effect of aspirin on the expression of ferritin, a cytoprotective protein that sequesters free cytosolic iron, the main catalyst of oxygen radical formation. In bovine pulmonary artery endothelial cells, aspirin at low antithrombotic concentrations (0.03 to 0.3 mmol/L) induced the synthesis of ferritin protein in a time- and concentration-dependent fashion up to 5-fold over basal levels, whereas ferritin H (heavy chain) mRNA remained unaltered. Aspirin-induced cytoprotection from hydrogen peroxide toxicity was mimicked by exogenous iron-free apoferritin but not iron-loaded ferritin, demonstrating the antioxidant function of newly synthesized ferritin under these conditions. Ferritin induction by aspirin was specific in that other nonsteroidal anti-inflammatory drugs such as salicylic acid, indomethacin, or diclofenac failed to alter ferritin protein levels. Aspirin-induced ferritin synthesis was abrogated in the presence of the iron chelator desferrioxamine, pointing to an interaction of aspirin with iron-responsive activation of ferritin translation. Together, our results suggest induction of ferritin as a novel mechanism by which aspirin may prevent endothelial injury in cardiovascular disease, eg, during atherogenesis.


Subject(s)
Antioxidants/metabolism , Aspirin/pharmacology , Endothelium, Vascular/metabolism , Ferritins/biosynthesis , Animals , Cattle , Cell Survival/drug effects , Cells, Cultured , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Gene Expression/drug effects , RNA, Messenger/genetics
10.
J Cell Biol ; 140(1): 211-21, 1998 Jan 12.
Article in English | MEDLINE | ID: mdl-9425168

ABSTRACT

Previously we have demonstrated that focal adhesion kinase (FAK)-promoted migration on fibronectin (FN) by its overexpression in CHO cells is dependent on FAK autophosphorylation at Y397 and subsequent binding of Src to this site. In this report, we have examined the role of FAK association with Grb2 and p130(Cas), two downstream events of the FAK/Src complex that could mediate integrin-stimulated activation of extracellular signal-regulated kinases (Erks). We show that a Y925F FAK mutant was able to promote cell migration as efficiently as FAK and that the transfected FAK demonstrated no detectable association with Grb2 in CHO cells. In contrast, cells expressing a FAK P712/715A mutant demonstrated a level of migration comparable to that of control cells. This mutation did not affect FAK kinase activity, autophosphorylation, or Src association but did significantly reduce p130(Cas) association with FAK. Furthermore, FAK expression in CHO cells increased tyrosine phosphorylation of p130(Cas) and its subsequent binding to several SH2 domains, which depended on both the p130(Cas) binding site and the Src binding site. However, we did not detect increased activation of Erks in cells expressing FAK, and the MEK inhibitor PD98059 did not decrease FAK-promoted cell migration. Finally, we show that coexpression of p130(Cas) further increased cell migration on FN and coexpression of the p130(Cas) SH3 domain alone functioned as a dominant negative mutant and decreased cell migration. Together, these results demonstrate that p130(Cas), but not Grb2, is a mediator of FAK-promoted cell migration and suggest that FAK/ p130(Cas) complex targets downstream pathways other than Erks in mediating FAK-promoted cell migration.


Subject(s)
Cell Adhesion Molecules/metabolism , Chemotaxis/physiology , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins , Animals , Binding Sites , CHO Cells , Cell Adhesion Molecules/biosynthesis , Cricetinae , Crk-Associated Substrate Protein , Focal Adhesion Protein-Tyrosine Kinases , Glutathione Transferase/biosynthesis , Mutagenesis, Site-Directed , Phosphoproteins/biosynthesis , Point Mutation , Polymerase Chain Reaction , Protein-Tyrosine Kinases/biosynthesis , Recombinant Fusion Proteins/metabolism , Retinoblastoma-Like Protein p130 , Transfection , src Homology Domains
11.
FEBS Lett ; 417(3): 349-51, 1997 Nov 17.
Article in English | MEDLINE | ID: mdl-9409749

ABSTRACT

A 24-h incubation with hydrogen peroxide (0.65 mM) markedly reduced viability of cultured endothelial cells. Preincubation with aspirin (3-30 microM) protected endothelial cells from hydrogen peroxide-induced toxicity and increased viability in a concentration-dependent fashion by up to 64% of control. A similar protection was observed with D-alpha-tocopherol acetate (vitamin E, 3-30 microM). The cytoprotective effects of aspirin and vitamin E against hydrogen peroxide were overadditive suggesting different mechanisms of antioxidant action. In agreement with this, cytotoxicity induced by iron, the main catalyst of oxygen radical formation, was substantially reduced by aspirin but not vitamin E. These results show that aspirin protects endothelial cells from oxidative stress possibly via binding or chelation of free cytosolic iron. Moreover, a combination of aspirin and vitamin E might be useful for the prevention of endothelial injury in cardiovascular disease, e.g. during atherogenesis.


Subject(s)
Aspirin/pharmacology , Cell Survival/drug effects , Endothelium, Vascular/physiology , Hydrogen Peroxide/toxicity , Oxidative Stress/physiology , Vitamin E/pharmacology , Animals , Cattle , Cell Line , Cytosol/metabolism , Drug Synergism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Iron/metabolism , Oxidative Stress/drug effects , Pulmonary Artery
12.
FEBS Lett ; 409(1): 46-8, 1997 Jun 02.
Article in English | MEDLINE | ID: mdl-9199501

ABSTRACT

In cultured endothelial cells, incubation with TNF-alpha (50 ng/ml) for 48 h markedly reduced viability of endothelial cells. A 6 h preincubation with Sper/NO (0.03--1 microM) protected endothelial cells in a concentration-dependent manner and increased viability by 63% of control. The NO scavenger PTIO (30 microM) completely abolished cytoprotection by Sper/NO. A cytoprotective effect comparable to Sper/NO was observed when preincubating the cells with 8-bromo cyclic GMP (1-10 microM). Moreover, no protection by Sper/NO occurred in the presence of ODQ (0.1 microM), a selective inhibitor of soluble guanylyl cyclase. Our results demonstrate that NO produces a long-term endothelial protection against cellular injury by TNF-alpha, presumably via a cyclic GMP-dependent pathway.


Subject(s)
Cyclic GMP/physiology , Cytotoxicity, Immunologic/drug effects , Endothelium, Vascular/drug effects , Nitric Oxide/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cattle , Cell Line , Cyclic GMP/immunology , Endothelium, Vascular/immunology , Pulmonary Artery/cytology , Spermine/pharmacology , Tumor Necrosis Factor-alpha/immunology
13.
Bioessays ; 19(2): 137-45, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9046243

ABSTRACT

Focal adhesion kinase (FAK) is a nonreceptor protein-tyrosine kinase implicated in controlling cellular responses to the engagement of cell-surface integrins, including cell spreading and migration, survival and proliferation. Aberrant FAK signaling may contribute to the process of cell transformation by certain oncoproteins, including v-Src. Progress toward elucidating the events leading to FAK activation following integrin-mediated cell adhesion, as well as events downstream of FAK, has come through the identification of FAK phosphorylation sites and interacting proteins. A signaling partnership is formed between FAK and Src-family kinases, leading to tyrosine phosphorylation of FAK and associated 'docking' proteins Cas and paxillin. Subsequent recruitment of proteins containing Src homology 2 domains, including Grb2 and c-Crk, to the complex is likely to trigger adhesion-induced cellular responses, including changes to the actin cytoskeleton and activation of the Ras-MAP kinase pathway.


Subject(s)
Cell Adhesion Molecules/physiology , Protein-Tyrosine Kinases/physiology , Signal Transduction , Animals , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans
14.
J Biol Chem ; 272(9): 5501-9, 1997 Feb 28.
Article in English | MEDLINE | ID: mdl-9038154

ABSTRACT

The focal adhesion kinase (FAK) and Crk-associated substrate, p130(Cas) (Cas), have been implicated in diverse signaling pathways including those mediated by integrins, G-protein-coupled receptors, tyrosine kinase receptors, and the v-src and v-crk oncogenes. The recent identification of a direct interaction between FAK and Cas prompted the examination of potential regulation of FAK.Cas complexes by factors that result in concomitant increase in their phosphotyrosine content, namely cell adhesion and transformation by Src. Both conditions resulted in elevated FAK.Cas complex levels in nonionic detergent-insoluble fractions, indicating increased association with the cytoskeleton. For activated Src, this effect requires an active Src catalytic domain but not its Src homology 2 (SH2) or Src homology 3 (SH3) domains. FAK kinase domain tyrosines 576 and 577 are also required, suggesting that direct phosphorylation of these sites by Src may influence the solubility and/or stability of the complex. FAK-Cas association was only observed in the context of Cas binding to at least one of two distinct proline-rich sites on FAK. These findings firmly establish a direct interaction between FAK and Cas and demonstrate that Src can influence the subcellular localization of the complex by a tyrosine phosphorylation-dependent mechanism.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion , Cytoskeleton/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Insulin/metabolism , Ubiquitin-Protein Ligases , src-Family Kinases/metabolism , Animals , COS Cells , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , Mice, Inbred BALB C , Octoxynol , Polyethylene Glycols , Proline , Proto-Oncogene Proteins c-cbl , Subcellular Fractions/metabolism , Tyrosine/metabolism
15.
J Mol Cell Cardiol ; 29(12): 3305-10, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9441836

ABSTRACT

In cultured endothelial cells, incubation with TNF-alpha (50 ng/ml) for 72 h markedly reduced viability of endothelial cells. A 6-h pre-incubation with the nitric oxide (NO) donor linsidomine (SIN-1, 10-150 microM) protected endothelial cells in a concentration-dependent manner and increased viability by up to 59% of control. The unmetabolized parent compound molsidomine and the NO-free metabolite of SIN-1 3-morpholinoiminoacetonitrile (SIN-1C) were without cytoprotective effect. Cytoprotection by SIN-1 was completely abolished by the NO scavenger 2-phenyl-4,4,5,5, -tetramethylimidazoline-1-oxyl-3-oxide (PTIO, 30 microM). A cytoprotective effect comparable to SIN-1 was observed when preincubating the cells with dibutyryl cyclic GMP (10-100 microM). Moreover, no protection by SIN-1 occurred in the presence of cycloheximide (1 microM) or 1H--1,2,4-oxadiazole-4, 3-a-quinoxalin-1-one (ODQ, 0.1 microM), a selective inhibitor of soluble guanylyl cyclase. Tin protoporphyrin-IX (SnPP, 25 microM), an inhibitor of heme oxygenase, was found to attenuate SIN-1-induced cytoprotection. Our results demonstrate that SIN-1 produces a long-term endothelial protection against cellular injury by TNF-alpha, presumably via a cyclic GMP-dependent pathway leading to up-regulation of protective proteins such as heme oxygenase.


Subject(s)
Cyclic GMP/metabolism , Endothelium, Vascular/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Molsidomine/analogs & derivatives , Nitric Oxide/metabolism , Tumor Necrosis Factor-alpha/toxicity , Vasodilator Agents/metabolism , Acetonitriles/metabolism , Animals , Cattle , Cells, Cultured , Drug Interactions , Endothelium, Vascular/drug effects , Molsidomine/metabolism , Morpholines/metabolism , Reactive Oxygen Species/metabolism
16.
Biochem Biophys Res Commun ; 228(3): 662-8, 1996 Nov 21.
Article in English | MEDLINE | ID: mdl-8941336

ABSTRACT

Focal adhesion kinase (FAK) participates in signaling events induced by diverse stimuli including integrin engagement, oncogenic transformation and mitogenic neuropeptides. FAK's signaling function is regulated by tyrosine phosphorylation. The major autophosphorylation site is tyrosine-397, which interacts with the Src homology 2 (SH2) domain of Src-family kinases including Src and Fyn. Full activation of FAK appears to require additional phosphorylation by the associated Src-family kinases. Previously identified Src sites include catalytic domain tyrosines-576 and -577, important for maximal FAK kinase activity, and tyrosine-925, which permits an SH2-mediated association with Grb2. A full understanding of FAK-mediated signaling events will require the identification of all sites of tyrosine phosphorylation. Here we report that tyrosine-861 is the major Src site in the carboxyl-terminal domain of FAK. Phosphotyrosine-861 may function in additional interactions between FAK and SH2-containing proteins.


Subject(s)
Cell Adhesion Molecules/metabolism , Protein-Tyrosine Kinases/metabolism , Tyrosine/metabolism , src Homology Domains , 3T3 Cells , Amino Acid Sequence , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics
17.
Proc Natl Acad Sci U S A ; 92(23): 10678-82, 1995 Nov 07.
Article in English | MEDLINE | ID: mdl-7479864

ABSTRACT

The focal adhesion kinase (FAK) has been implicated in integrin-mediated signaling events and in the mechanism of cell transformation by the v-Src and v-Crk oncoproteins. To gain further insight into FAK signaling pathways, we used a two-hybrid screen to identify proteins that interact with mouse FAK. The screen identified two proteins that interact with FAK via their Src homology 3 (SH3) domains: a v-Crk-associated tyrosine kinase substrate (Cas), p130Cas, and a still uncharacterized protein, FIPSH3-2, which contains an SH3 domain closely related to that of p130Cas. These SH3 domains bind to the same proline-rich region of FAK (APPKPSR) encompassing residues 711-717. The mouse p130Cas amino acid sequence was deduced from cDNA clones, revealing an overall high degree of similarity to the recently reported rat sequence. Coimmunoprecipitation experiments confirmed that p130Cas and FAK are associated in mouse fibroblasts. The stable interaction between p130Cas and FAK emerges as a likely key element in integrin-mediated signal transduction and further represents a direct molecular link between the v-Src and v-Crk oncoproteins. The Src family kinase Fyn, whose Src homology 2 (SH2) domain binds to the major FAK autophosphorylation site (tyrosine 397), was also identified in the two-hybrid screen.


Subject(s)
Cell Adhesion Molecules/metabolism , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins , Signal Transduction , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , Crk-Associated Substrate Protein , DNA, Complementary/genetics , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphoproteins/genetics , Precipitin Tests , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-crk , Retinoblastoma-Like Protein p130 , Sequence Analysis, DNA , Sequence Homology, Amino Acid , src Homology Domains
18.
Mol Cell Biol ; 15(2): 954-63, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7529876

ABSTRACT

Focal adhesion kinase (FAK) is a widely expressed nonreceptor protein-tyrosine kinase implicated in integrin-mediated signal transduction pathways and in the process of oncogenic transformation by v-Src. Elevation of FAK's phosphotyrosine content, following both cell adhesion to extracellular matrix substrata and cell transformation by Rous sarcoma virus, correlates directly with an increased kinase activity. To help elucidate the role of FAK phosphorylation in signal transduction events, we used a tryptic phosphopeptide mapping approach to identify tyrosine sites of phosphorylation responsive to both cell adhesion and Src transformation. We have identified four tyrosines, 397, 407, 576, and 577, which are phosphorylated in mouse BALB/3T3 fibroblasts in an adhesion-dependent manner. Tyrosine 397 has been previously recognized as the major site of FAK autophosphorylation. Phosphorylation of tyrosines 407, 576, and 577, which are previously unrecognized sites, is significantly elevated in the presence of c-Src in vitro and v-Src in vivo. Tyrosines 576 and 577 lie within catalytic subdomain VIII--a region recognized as a target for phosphorylation-mediated regulation of protein kinase activity. We found that maximal kinase activity of FAK immune complexes requires phosphorylation of both tyrosines 576 and 577. Our results indicate that phosphorylation of FAK by Src (or other Src family kinases) is an important step in the formation of an active signaling complex.


Subject(s)
Cell Adhesion Molecules/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Tyrosine/analogs & derivatives , 3T3 Cells , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Avian Sarcoma Viruses , Base Sequence , Binding Sites , Cell Adhesion , Cell Transformation, Neoplastic , Chlorocebus aethiops , Electrophoresis, Gel, Two-Dimensional , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Peptide Mapping , Phosphopeptides/chemistry , Phosphopeptides/isolation & purification , Phosphorylation , Phosphotyrosine , Receptor, Insulin/metabolism , Recombinant Proteins , Transfection , Tyrosine/analysis , Tyrosine/metabolism
19.
J Cell Biochem ; 55(1): 106-19, 1994 May.
Article in English | MEDLINE | ID: mdl-7521880

ABSTRACT

Focal adhesion kinase (FAK) is a structurally unique nonreceptor protein-tyrosine kinase that localizes to focal adhesion plaques. Regulation of its activity has been implicated in diverse signaling pathways, including those mediated by extracellular matrix/integrin interactions, G-protein coupled receptors for mitogenic neuropeptides, and certain oncogene products. To gain evidence for specific processes in which FAK may be involved in vivo, a study was initiated to determine its expression pattern during mouse development. FAK expression was detected in early embryos and appeared to be distributed throughout all cell types at about the time of neurulation. Subsequent to neural tube closure, expression became particularly abundant in the developing vasculature. This included expression in the medial layer of arteries populated by smooth muscle cells. In vitro studies using cultured rat aortic vascular smooth muscle cells demonstrate that FAK phosphotyrosine content is dramatically elevated in response to plating cells onto the adhesive glycoprotein, fibronectin. Also, enhanced tyrosine phosphorylation of FAK is observed in these cells upon stimulation with the vasoconstrictor angiotensin II. Thus, in vascular smooth muscle cells, like fibroblasts, FAK appears to play a role in signaling mechanisms induced by extracellular matrix components as well as G-protein coupled receptor agonists. The combined results of this study suggest that signaling through FAK may play an important role in blood vessel morphogenesis and function.


Subject(s)
Angiotensin II/pharmacology , Cell Adhesion Molecules/biosynthesis , Muscle, Smooth, Vascular/enzymology , Protein-Tyrosine Kinases/biosynthesis , Tyrosine/analogs & derivatives , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Embryonic and Fetal Development/physiology , Enzyme Induction/drug effects , Female , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/embryology , Phosphotyrosine , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Time Factors , Tyrosine/metabolism
20.
Biochem Biophys Res Commun ; 190(3): 1084-9, 1993 Feb 15.
Article in English | MEDLINE | ID: mdl-8439308

ABSTRACT

A novel protein-tyrosine kinase, designated 'Focal Adhesion Kinase' (FAK), has recently been implicated in signal transduction pathways activated by extracellular adhesion molecules and by neuropeptide growth factors. Previously deduced primary structures for chicken and mouse FAK polypeptides differ at their amino-termini, with mouse FAK reported to have a 25 amino acid residue extension not present in chicken FAK. Additional sequence information from the 5'-end region of the chicken FAK transcript now indicates that the amino-terminal extension previously thought to be unique to mouse FAK is, in fact, also predicted for chicken FAK. Thus mouse and chicken FAK polypeptides appear to be structurally similar throughout their lengths. This is further supported by comparison of their electrophoretic mobilities.


Subject(s)
Cell Adhesion Molecules/chemistry , Protein-Tyrosine Kinases/chemistry , Amino Acid Sequence , Animals , Base Sequence , Chick Embryo , DNA/genetics , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/genetics , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...