Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Ultramicroscopy ; 107(10-11): 989-94, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17644253

ABSTRACT

In this work, we propose a method to estimate basic parameters like the rms roughness and the mean grain size of nanocrystalline thin films on rough substrates. The method is based on the analysis of the power spectral density (PSD) of the surface profile, which allows distinguishing between the two participating components from surface and film. The effectiveness will be demonstrated for thin NiO(x) layers for gas sensing on Al(2)O(3) ceramic substrates, and for protective WC coatings on steel.

2.
Ultramicroscopy ; 107(10-11): 1086-90, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17587498

ABSTRACT

Knowledge of tip geometry is necessary for reproducible atomic force microscope (AFM) measurements. This is particularly important for measurements in contact mode, in which a certain wear of the tip will always occur. For small or flat structures or for structures of larger dimensions, knowledge of the tip radius and the entire tip geometry is important. Additionally, the tilt of the tip in relation to the sample is of importance. Normally, very complicated lithographically manufactured structures for tip characterization are used. In contrast, the structures shown in this work are very simple. For measuring the tip geometry very thin foils patterned by focused ion beam (FIB) were used. In this work we demonstrate the possibility of determining the AFM tip geometry and the tilt based on several different large structures. A proven algorithm was developed for the reconstruction of the tips. The shape of FIB-structured foils was determined by electron microscopy prior to AFM measurements. This new method for determining tip shape is also presented as it compares to other current methods. In this case a discussion on the stability and advantages of the new method is presented.

3.
Infect Immun ; 69(2): 897-905, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11159983

ABSTRACT

The interaction of Listeria monocytogenes with endothelial cells represents a crucial step in the pathogenesis of listeriosis. Incubation of human umbilical vein endothelial cells (HUVEC) with wild-type L. monocytogenes (EGD) provoked immediate strong NO synthesis, attributable to listerial presentation of listeriolysin O (LLO), as the NO release was missed upon employment of a deletion mutant for LLO (EGD hly mutant) and was reproduced by purified LLO. Studies of conditions lacking extracellular Ca(2+) suggested LLO-elicited Ca(2+) flux as the underlying mechanism. In addition, HUVEC incubation with EGD turned out to be a potent stimulus for sustained (>12-h) upregulation of proinflammatory cytokine generation (interleukin 6 [IL-6], IL-8, and granulocyte-macrophage colony-stimulating factor). Use of deletion mutants for LLO (EGD hly mutant), listerial phosphatidylinositol-specific phospholipase C (EGD plcA mutant), broad-spectrum phospholipase C (EGD plcB mutant) and internalin B (EGD inlB mutant), as well as purified LLO, identified LLO as largely responsible for the cytokine response. Endothelial cells responded with diacylglycerole and ceramide generation as well as nuclear translocation of NF-kappa B to the stimulation with the LLO-producing strains EGD and Listeria innocua. The endothelial PC-phospholipase C inhibitor tricyclodecan-9-yl-xanthogenate as well as two independent inhibitors of NF-kappa B activation, pyrolidine dithiocarbamate and caffeic acid phenethyl ester, suppressed both the NF-kappa B translocation and the upregulation of cytokine synthesis. We conclude that L. monocytogenes is a potent stimulus of NO release and sustained upregulation of proinflammatory cytokine synthesis in human endothelial cells, both events being largely attributable to LLO presentation. LLO-induced transmembrane Ca(2+) flux as well as a sequence of endothelial phospholipase activation and the appearance of diacylglycerole, ceramide, and NF-kappa B are suggested as underlying host signaling events. These endothelial responses to L. monocytogenes may well contribute to the pathogenic sequelae in severe listerial infection and sepsis.


Subject(s)
Bacterial Toxins , Endothelium, Vascular/metabolism , Heat-Shock Proteins/toxicity , Inflammation Mediators/metabolism , Listeria monocytogenes/pathogenicity , Calcium/metabolism , Cells, Cultured , Ceramides/metabolism , Diglycerides/metabolism , Endothelium, Vascular/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hemolysin Proteins , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Virulence
4.
J Cell Biol ; 148(1): 17-28, 2000 Jan 10.
Article in English | MEDLINE | ID: mdl-10629215

ABSTRACT

Caveolin-1 is an integral membrane protein of caveolae that is thought to play an important role in both the traffic of cholesterol to caveolae and modulating the activity of multiple signaling molecules at this site. The molecule is synthesized in the endoplasmic reticulum, transported to the cell surface, and undergoes a poorly understood recycling itinerary. We have used mutagenesis to determine the parts of the molecule that control traffic of caveolin-1 from its site of synthesis to the cell surface. We identified four regions of the molecule that appear to influence caveolin-1 traffic. A region between amino acids 66 and 70, which is in the most conserved region of the molecule, is necessary for exit from the endoplasmic reticulum. The region between amino acids 71 and 80 controls incorporation of caveolin-1 oligomers into detergent-resistant regions of the Golgi apparatus. Amino acids 91-100 and 134-154 both control oligomerization and exit from the Golgi apparatus. Removal of other portions of the molecule has no effect on targeting of newly synthesized caveolin-1 to caveolae. The results suggest that movement of caveolin-1 among various endomembrane compartments is controlled at multiple steps.


Subject(s)
Caveolins , Membrane Proteins/metabolism , Animals , Binding Sites , Biological Transport , CHO Cells , Caveolin 1 , Cricetinae , Intracellular Fluid/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutagenesis
5.
Nat Cell Biol ; 1(6): 369-75, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10559965

ABSTRACT

Caveolin-1 is a protein component (of relative molecular mass 22, 000) of the striated coat that decorates the cytoplasmic surface of caveolae membranes. Previous biochemical and molecular tests have indicated that caveolin-1 is an integral membrane protein that is co-translationally inserted into endoplasmic-reticulum membranes of fibroblast and epithelial cells such that its carboxy- and amino-terminal ends are in the cytoplasm. Here we identify caveolin-1 in the secretory pathway of exocrine cells. Secretion of caveolin-1 from pancreatic acinar cells and a transfected exocrine cell line, but not from Chinese hamster ovary cells, is stimulated by the secretagogues secretin, cholecystokinin and dexamethasone. The secreted caveolin-1 co-fractionates with apolipoproteins, indicating that it may be secreted in a complex with lipids.


Subject(s)
Apolipoproteins/metabolism , Caveolins , Membrane Proteins/metabolism , Pancreas/metabolism , Animals , Apolipoproteins/chemistry , CHO Cells , Caveolin 1 , Cricetinae , Humans , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Pancreas/cytology , Rats , Rats, Sprague-Dawley , Transfection
6.
J Biol Chem ; 274(15): 10203-12, 1999 Apr 09.
Article in English | MEDLINE | ID: mdl-10187805

ABSTRACT

The 55-kDa receptor for tumor necrosis factor (TR55) triggers multiple signaling cascades initiated by adapter proteins like TRADD and FAN. By use of the primary amine monodansylcadaverine (MDC), we addressed the functional role of tumor necrosis factor (TNF) receptor internalization for intracellular signal distribution. We show that MDC does not prevent the interaction of the p55 TNF receptor (TR55) with FAN and TRADD. Furthermore, the activation of plasmamembrane-associated neutral sphingomyelinase activation as well as the stimulation of proline-directed protein kinases were not affected in MDC-treated cells. In contrast, activation of signaling enzymes that are linked to the "death domain" of TR55, like acid sphingomyelinase and c-Jun-N-terminal protein kinase as well as TNF signaling of apoptosis in U937 and L929 cells, are blocked in the presence of MDC. The results of our study suggest a role of TR55 internalization for the activation of select TR55 death domain signaling pathways including those leading to apoptosis.


Subject(s)
Antigens, CD/metabolism , Apoptosis , Cadaverine/analogs & derivatives , Mitogen-Activated Protein Kinases , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Cadaverine/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Ceramides/metabolism , Endocytosis , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Potassium/metabolism , Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I , Sphingomyelin Phosphodiesterase/metabolism , TNF Receptor-Associated Factor 1 , U937 Cells , fas Receptor/immunology , fas Receptor/metabolism
7.
FEBS Lett ; 436(1): 51-4, 1998 Sep 25.
Article in English | MEDLINE | ID: mdl-9771892

ABSTRACT

Kinectin has been characterized as the first known receptor for the molecular motor kinesin, which is critically involved in microtubule-based vesicle transport and membrane trafficking. Here we identify kinectin as a target for caspase-mediated proteolysis during apoptosis. Treatment of cells with diverse apoptotic stimuli including TNF, anti-Fas, anticancer drugs, gamma-radiation or ceramide leads to rapid proteolytic cleavage of the 160-kDa form of kinectin to a 120-kDa fragment. Evidence is provided that kinectin cleavage is mediated by caspase 7.


Subject(s)
Caspases/metabolism , Membrane Proteins , Receptors, Cell Surface/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Antibodies/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase 7 , Caspases/drug effects , Caspases/genetics , Cell-Free System , Ceramides/pharmacology , Cisplatin/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Daunorubicin/pharmacology , Etoposide/pharmacology , HeLa Cells/drug effects , HeLa Cells/metabolism , HeLa Cells/radiation effects , Humans , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Jurkat Cells/radiation effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , fas Receptor/immunology , fas Receptor/metabolism
8.
Transplantation ; 66(12): 1818-22, 1998 Dec 27.
Article in English | MEDLINE | ID: mdl-9884281

ABSTRACT

BACKGROUND: Soluble MHC class I molecules are ubiquitous in human body fluids, including serum, urine, sweat, and cerebrospinal fluid. However, their biological function has remained unresolved. Membrane-derived human soluble MHC molecules (soluble human leukocyte antigen; sHLA) have been shown to induce apoptosis in alloreactive cytotoxic T lymphocytes (CTL). Here we report the efficacy of recombinant soluble HLA-B7 (rsHLA-B7) to modulate T-cell function. METHODS: Primers of HLA-B7 were designed to allow amplification of a cDNA lacking the transmembrane and cytoplasmic domains yielding a truncated gene. rsHLA-B7 molecules were expressed in the human myeloma cell line 721.221 and purified by affinity chromatography using the BB7.7 mouse monoclonal antibody. CTL were generated from peripheral blood lymphocytes derived from healthy blood donors by stimulation with irradiated Epstein Barr virus-transformed HLA-B7-positive B cells. CTL were preincubated with rsHLA-B7, and cytotoxicity and apoptosis were tested according to standard procedure. RESULTS: A total of 2 x 10(6) cells/ml secreted 10 microg/ml rsHLA-B7 as determined by a conformation-dependent ELISA, suggesting that rsHLA-B7 do not require the transmembrane and cytoplasmic regions for proper folding. After purification by affinity chromatography, rsHLA-B7 induced apoptosis in anti-HLA-B7 CTL, but not in anti-HLA-A2-specific, CTL. As a consequence, allorecognition of target cells by the CTL was significantly blocked. CONCLUSION: Recombinant sHLA are sufficient binding cues for T cells, which efficiently induce apoptosis and block allorecognition of target cells by CTL. Thus, recombinant sHLA molecules may become a valuable new modality for specific immunological therapeutic intervention.


Subject(s)
Apoptosis/drug effects , HLA-B7 Antigen/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , Animals , HLA-B7 Antigen/chemistry , Humans , Mice , Protein Folding , Recombinant Proteins/pharmacology , T-Lymphocytes, Cytotoxic/physiology
9.
J Exp Med ; 184(2): 725-33, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8760826

ABSTRACT

Tumor necrosis factor (TNF) is a pleiotropic mediator of inflammation that has been implicated in the pathogenesis of devastating clinical syndromes including septic shock. We have investigated the role of a TNF-responsive phosphatidylcholine-specific phospholipase C (PC-PLC) for the cytotoxic and proinflammatory activity of TNF. We show here that the cytotoxicity signaled for by the so-called "death domain" of the p55 TNF receptor is associated with the activation of PC-PLC. The xanthogenate tricyclodecan-9-yl (D609), a specific and selective inhibitor of PC-PLC, blocked the cytotoxic action of TNF on L929 and Wehi164 cells. In vivo, D609 prevented both adhesion molecule expression in the pulmonary vasculature and the accompanying leukocyte infiltration in TNF-treated mice. More strikingly, D609 protects BALB/c mice from lethal shock induced either by TNF, lipopolysaccharide, or staphylococcal enterotoxin B. Together these findings imply PC-PLC as an important mediator of the pathogenic action of TNF, suggesting that PC-PLC may serve as a novel target for anti-inflammatory TNF antagonists.


Subject(s)
Antigens, CD/physiology , Phosphatidylcholines/physiology , Receptors, Tumor Necrosis Factor/physiology , Type C Phospholipases/physiology , Animals , Bridged-Ring Compounds/pharmacology , Cell Line , Enterotoxins , Enzyme Activation , Mice , Norbornanes , Phosphodiesterase Inhibitors/pharmacology , Phospholipases A/metabolism , Receptors, Tumor Necrosis Factor, Type I , Shock, Septic/prevention & control , Signal Transduction , Thiocarbamates , Thiones/pharmacology , Type C Phospholipases/antagonists & inhibitors
10.
J Immunol ; 156(4): 1587-93, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8568264

ABSTRACT

Stimulation of leukocyte adhesion to the endothelium by TNF is mediated by the up-regulation of adhesion molecules on the endothelial cell surface. C57BL/6 mice and syngenic 55-kDa TNF receptor-deficient mice (TNFRp55-/- mice) were challenged with TNF, and the kinetics of intracellular adhesion molecule-1, ICAM-1, mucosal addressin cell adhesion molecule-1, vascular adhesion molecule-1 (VCAM-1), and E-selectin expression were examined in various organs. TNF induced sustained VCAM-1 expression within 4 h in lung, liver, and kidney. In the lungs, but not in other organs, transient E-selectin expression was induced by TNF within 0.5 h and peaked at 4 h. The TNF-induced expression of VCAM-1 and E-selectin was found to be exclusively controlled by the 55-kDa TNF-receptor (TNFRp55) as demonstrated by analysis of TNFRp55-/- mice. Furthermore, TNF triggered mononuclear cell and neutrophil infiltration of lung, liver, and kidney in C57BL/6 mice but not TNFRp55-/- mice. Interestingly, MAdCAM-1 expression in the marginal sinus of the spleen was detected in wild-type mice but was absent in TNFRp55-/- mice. Together, the data suggest that in vivo the 55-kDa TNF receptor mediates the induction of VCAM-1 and E-selectin expression and is critically involved in the control of leukocyte organ infiltration.


Subject(s)
E-Selectin/metabolism , Leukocytes/cytology , Receptors, Tumor Necrosis Factor/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Cell Adhesion , Endothelium, Vascular/cytology , Female , Immunoenzyme Techniques , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/metabolism , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Immunobiology ; 193(2-4): 193-203, 1995 Jul.
Article in English | MEDLINE | ID: mdl-8530143

ABSTRACT

Tumor Necrosis Factor (TNF) is one of the most potent physiological inducers of the nuclear transcription factor NF-kappa B. In light of the pivotal role of NF-kappa B in the development of immune responses and activation of HIV replication, the identification of TNF signal transduction pathways involved in NF-kappa B activation is of particular interest. Data from our laboratory demonstrate that the TNF signal transduction pathway-mediating NF-kappa B activation involves two phospholipases, a phosphatidylcholine-specific phospholipase C (PC-PLC) and an endosomal acidic sphingomyelinase (aSMase). The aSMase activation by TNF is secondary to the generation of 1,2-diacylglycerol (DAG) produced by a TNF-responsive PC-PLC. SMase and its product ceramide induce degradation of the NF-kappa B inhibitor I kappa B as well as NF-kappa B activation. Besides endosomal acidic SMase, TNF also rapidly activates a plasmamembrane-associated neural SMase (nSMase), that, however is not involved in TNF-induced NF-kappa B activation. NSMase and aSMase are activated by different cytoplasmic domains of the 55 kDa TNF-receptor and are coupled to select pathways of TNF signaling. Ceramide generated by nSMase directs the activation of proline-directed serin/threonine protein kinases and phospholipase A2 and ceramide produced by aSMase triggers the activation of NF-kappa B. No apparent crosstalk was detected between nSMase and aSMase pathways, indicating that ceramide action depends on the topology of its production.


Subject(s)
NF-kappa B/biosynthesis , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/physiology , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , NF-kappa B/drug effects , Signal Transduction/drug effects
12.
J Exp Med ; 181(6): 2059-68, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7759998

ABSTRACT

T cell receptor recognition of antigen can lead either to T lymphocyte differentiation and proliferation or to a state of unresponsiveness, which is dependent on whether appropriate costimulatory signals are provided to the mature T cell. We have investigated a novel intracellular signaling pathway provided by the costimulatory molecule CD28. CD28 engagement triggers the activation of an acidic sphingomyelinase (A-SMase), which results in the generation of ceramide, an important lipid messenger intermediate. A-SMase activation by CD28 occurred in resting as well as in activated primary T cells or leukemic Jurkat cells. In contrast, ligation of either CD3 or CD2 did not result in A-SMase activation. Overexpression of recombinant A-SMase in Jurkat T cells substituted for CD28 with regard to nuclear factor-kB activation. These data suggest that CD28 provides an important costimulatory signal by activation of an acidic sphingomyelinase pathway.


Subject(s)
CD28 Antigens/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes/physiology , Animals , Cell Differentiation , Cell Line , Cricetinae , Enzyme Activation , Female , Humans , Kinetics , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Phytohemagglutinins , T-Lymphocytes/cytology , T-Lymphocytes/enzymology
13.
J Inflamm ; 45(3): 183-92, 1995.
Article in English | MEDLINE | ID: mdl-8597873

ABSTRACT

Superantigens, such as staphylococcal enterotoxins (SE), cause food poisoning and shock, which is mediated at least in part by TNF. We have examined the mechanism of superantigen-induced activation of the TNF gene promoter in primary human peripheral blood T lymphocytes and in the leukemic T cell line Jurkat. Like stimulation of the T cell receptor complex through CD3 ligation, SEB induces binding of nuclear proteins to Egr- and Jun/ATF-related consensus sequences present between nucleotides -170 and -100 of the TNF promoter 5' flanking region. By cotransfection of Jurkat T cells with constructs containing TNF promoter deletion mutants linked to a CAT reporter gene, it is shown that superantigens can activate transcription from these two adjacent ETs and Jun/ATF binding elements. Superantigen-induced binding of Egr-1 and Jun/ATF is markedly reduced by okadaic acid, suggesting that phosphatases are involved in the signaling of SE. When compared to CD3 ligation, superantigens activate the TNF promoter more potently, which is likely due to costimulatory signals provided by superantigen presenting cells.


Subject(s)
Promoter Regions, Genetic , Superantigens/pharmacology , T-Lymphocytes/metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics , Animals , Chloramphenicol O-Acetyltransferase/genetics , Enterotoxins/pharmacology , Gene Deletion , Humans , Leukemia , Mice , Transfection , Tumor Cells, Cultured
14.
J Leukoc Biol ; 56(5): 533-41, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7964160

ABSTRACT

Tumor necrosis factor (TNF) and interleukin-1 (IL-1) are cytokines with pleiotropic biological activities, exerting a broad range of overlapping biological functions. The redundancy of TNF and IL-1 activities may be based on the utilization of shared key components of intracellular signaling pathways. Two lipid second messengers have been found to transmit TNF and IL-1 intracellular signals: 1,2-diacylglycerol (DAG), generated by a phosphatidylcholine-specific phospholipase C, and ceramide, generated by sphingomyelinase (SMase). DAG is a well established activator of the important signaling system protein kinase C (PKC), which appears to mediate various cellular responses to TNF or IL-1. In addition, it is obvious that DAG also activates other enzyme systems like acidic sphingomyelinase. SMases have been implicated in a number of TNF responses, including stimulation of cell growth and differentiation, as well as triggering cytotoxicity and apoptosis. The metabolic active cleavage product of SMase, ceramide, is a novel multifunctional lipid second messenger capable of inducing various signaling systems. Both cytokines, TNF and IL-1, stimulate a neutral,plasma membrane-associated SMase that leads to stimulation of a protein kinase and eventually to activation of the mitogen-activated protein (MAP) kinase cascade and phospholipase A2. Ceramide is also capable of stimulating a cytosolic protein phosphatase. PKC plays a role in activation of the nuclear transcription factor AP-1, and the DAG-regulated acidic SMase is involved in transducing TNF signals to the cell nucleus via activation of the nuclear transcription factor NF-kappa B.


Subject(s)
Ceramides/biosynthesis , Diglycerides/biosynthesis , Interleukin-1/physiology , Protein Kinase C/physiology , Second Messenger Systems/physiology , Sphingomyelin Phosphodiesterase/metabolism , Tumor Necrosis Factor-alpha/physiology , Type C Phospholipases/metabolism , Animals , Enzyme Activation , Humans , NF-kappa B/metabolism
15.
Cell ; 78(6): 1005-15, 1994 Sep 23.
Article in English | MEDLINE | ID: mdl-7923351

ABSTRACT

Ceramide produced by sphingomyelinases (SMases) has been recognized as an important second messenger in growth factor receptor signaling. Tumor necrosis factor (TNF), through binding to the 55 kDa TNF receptor (TNF-R55), rapidly activates two distinct types of SMase, a membrane-associated neutral (N-)SMase, and an endosomal acidic (A)-SMase. N-SMase and A-SMase are activated independently by different cytoplasmic domains of TNF-R55. Each type of SMase specifically couples to select pathways of TNF signaling. Ceramide generated by N-SMase directs the activation of proline-directed serine/threonine protein kinase(s) and phospholipase A2. In contrast, A-SMase triggers the activation of NF-kappa B. No apparent crosstalk was detected between N-SMase and A-SMase pathways, indicating that ceramide action depends on the topology of its production. These results suggest that N-SMase and A-SMase control important yet dissociable and nonoverlapping pathways of TNF receptor signal transduction.


Subject(s)
Isoenzymes/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/physiology , Sphingomyelin Phosphodiesterase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Base Sequence , Bridged-Ring Compounds/pharmacology , Ceramides/metabolism , Enzyme Activation , Humans , Hydrogen-Ion Concentration , Mice , Models, Biological , Molecular Sequence Data , NF-kappa B/analysis , Norbornanes , Phospholipases A/analysis , Phospholipases A2 , Protein Kinases/analysis , Receptors, Tumor Necrosis Factor/genetics , Recombinant Proteins/metabolism , Second Messenger Systems/physiology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Thiocarbamates , Thiones/pharmacology , Transfection , Tumor Cells, Cultured , Type C Phospholipases/analysis
16.
J Biol Chem ; 269(19): 13760-5, 1994 May 13.
Article in English | MEDLINE | ID: mdl-8188652

ABSTRACT

Tumor necrosis factor (TNF) is one of the most potent physiological inducers of the nuclear transcription factor kappa B (NF-kappa B). A key event in the activation of NF-kappa B is the rapid release of the inhibitory subunit I kappa B-alpha. Various inhibitors of serine-like proteases are shown to block TNF-mediated NF-kappa B activation as well as the disappearance of I kappa B-alpha immunoreactivity in primary murine T lymphocytes and in various human leukemic cell lines. The protease inhibitors did not block TNF-induced activation of either phosphatidylcholine-specific phospholipase C or acidic sphingomyelinase (SMase), indicating that the putative protease operates rather downstream of TNF signal transduction processes. I kappa B-alpha degradation could be directly induced by addition of sphingomyelinase or synthetic ceramide to a cell-free system, indicating a stringent coupling of SMase to the NF-kappa B activation pathway. SMase-induced I kappa B-alpha degradation was suppressed by the protease inhibitor dichloroisocoumarin. Together, the data suggest that a TNF-responsive sphingomyelinase triggers the rapid degradation of I kappa B-alpha through a serine-like protease, which appears to be crucial to the control of NF-kappa B activation.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Proteins , Sphingomyelin Phosphodiesterase/metabolism , Animals , Base Sequence , Cell Line , Cell-Free System , Humans , Hydrolysis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , NF-KappaB Inhibitor alpha , Oligodeoxyribonucleotides , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
17.
Nature ; 365(6442): 182-5, 1993 Sep 09.
Article in English | MEDLINE | ID: mdl-8371761

ABSTRACT

Inducible gene expression in eukaryotes is mainly controlled by the activity of transcriptional activator proteins, such as NF-kappa B (refs 1-3), a factor activated upon treatment of cells with phorbol esters, lipopolysaccharide, interleukin-1 and tumour necrosis factor-alpha. Activation of NF-kappa B involves release of the inhibitory subunit I kappa B from a cytoplasmic complex with the DNA-binding subunits Rel-A (formerly p65) and p50 (refs 6, 7). Cell-free experiments have suggested that protein kinase C and other kinases transfer phosphoryl groups onto I kappa B causing release of I kappa B and subsequent activation of NF-kappa B. Here we report that I kappa B-alpha (formerly MAD-3) is degraded in cells after stimulation with phorbol ester, interleukin-1, lipopolysaccharide and tumour necrosis factor-alpha, an event coincident with the appearance of active NF-kappa B. Treatment of cells with various protease inhibitors or an antioxidant completely prevented the inducible decay of I kappa B-alpha as well as the activation of NF-kappa B. Our findings suggest that the activation of NF-kappa B relies on an inducible degradation of I kappa B-alpha through a cytoplasmic, chymotrypsin-like protease. In intact cells, phosphorylation of I kappa B-alpha is apparently not sufficient for activation of NF-kappa B.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Proteins , NF-kappa B/metabolism , B-Lymphocytes/metabolism , Blotting, Western , Cell Line , Cycloheximide/pharmacology , DNA/metabolism , Endopeptidases/metabolism , HeLa Cells , Humans , Hydrolysis , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , NF-KappaB Inhibitor alpha , Protease Inhibitors/pharmacology , Protein Binding/drug effects , Reactive Oxygen Species/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
18.
Cell ; 71(5): 765-76, 1992 Nov 27.
Article in English | MEDLINE | ID: mdl-1330325

ABSTRACT

In this paper, we describe a phospholipid transmission pathway mediating tumor necrosis factor (TNF) activation of the nuclear transcription factor kappa B (NF-kappa B). Central to this TNF signaling route is the second messenger-like molecule ceramide, which is generated by sphingomyelin (SM) breakdown catalyzed by a sphingomyelinase (SMase). SMase activation is secondary to the generation of 1,2-diacylglycerol (DAG) produced by a TNF-responsive PC-specific phospholipase C (PC-PLC). The functional coupling of these two C type phospholipases is revealed by D609, a selective inhibitor of PC-PLC. SMase itself, or SMase-inducing regimens such as exogenous PLC or synthetic DAGs, induces NF-kappa B activation at pH 5.0, suggesting the operation of an acidic SMase. A model is proposed in which a TNF-responsive PC-PLC via DAG couples to an acidic SMase, resulting in the generation of ceramide, which eventually triggers rapid induction of nuclear NF-kappa B activity.


Subject(s)
NF-kappa B/metabolism , Phosphatidylcholines/metabolism , Sphingomyelins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Type C Phospholipases/physiology , Base Sequence , Ceramides/pharmacology , Diglycerides/pharmacology , Isoelectric Point , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Receptors, Cell Surface/physiology , Receptors, Tumor Necrosis Factor , Regulatory Sequences, Nucleic Acid , Signal Transduction , Sphingomyelin Phosphodiesterase/metabolism
19.
J Biol Chem ; 267(25): 17997-8001, 1992 Sep 05.
Article in English | MEDLINE | ID: mdl-1325455

ABSTRACT

The numerous biological activities of tumor necrosis factor (TNF) appear mediated by two types of receptors of 55 kDa (TR55) and 75 kDa (TR75) molecular mass. To test TR55 for its individual role in signaling across the membrane, a cDNA coding for the human TR55 was stably expressed in murine 70Z/3 pre-B cells, which lack binding sites for, and proved nonresponsive to human TNF. The transfected TR55 showed high affinity ligand binding and active internalization. It is demonstrated that the TNF signaling cascade, i.e. stimulation of protein kinase C, sphingomyelinase, and phospholipase A2, production of the second messengers diacylglycerol and ceramide, can occur completely through exclusive binding of TNF to TR55. The p55 TNF-binding site functions as an autonomous TNF receptor that mediates key signal transduction pathways, which may control the majority of TNF actions.


Subject(s)
Receptors, Cell Surface/physiology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , Base Sequence , Cell Line , Cell Nucleus/metabolism , Ceramides/metabolism , Diglycerides/metabolism , Enhancer Elements, Genetic , HIV Long Terminal Repeat , HIV-1/genetics , Humans , Kinetics , L Cells , Mice , Molecular Sequence Data , Molecular Weight , NF-kappa B/metabolism , Oligodeoxyribonucleotides , Phospholipases A/metabolism , Phospholipases A2 , Polymerase Chain Reaction , Protein Kinase C/metabolism , Radioligand Assay , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor , Second Messenger Systems/drug effects , Signal Transduction/drug effects , Transfection , Tumor Necrosis Factor-alpha/pharmacology
20.
J Immunol ; 148(10): 3152-7, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1315830

ABSTRACT

The numerous biologic activities of TNF appear mediated by two types of specific cell surface receptors of 55 to 60 kDa (TR55) and 75 to 80 kDa (TR75) molecular mass, respectively. The role of TR55 in the activation of the nuclear transcription factor kappa B (NF-kappa B) was investigated using an antagonistic, mAb, H398, specific for the human TR55. The human leukemic T cell line, Jurkat, which expresses both types of receptors at comparable levels, was used to test for NF-kappa B activation by electrophoretic mobility shift assays using as a probe an oligonucleotide encompassing the two tandemly arranged kappa B sites of the HIV-1 LTR enhancer. mAb H398 is shown to efficiently block not only TNF- but also lymphotoxin-mediated activation of NF-kappa B. Furthermore mAb H398 also impeded TNF- or lymphotoxin-mediated activation of chloramphenicol acetyl transferase gene expression from the HIV-1-LTR as determined by transient transfection assays. These findings indicate that both, induction of NF-kappa B binding to DNA, and transcriptional activity can be efficiently inhibited by selective blockade of TR55. Finally it is shown, that human TR55 confers NF-kappa B inducibility when expressed in the mouse pre-B cell line 70Z/3, which does not respond to TNF in its parental state. Together, the results of this study indicate that TR55 is both necessary and sufficient for mediating TNF activation of NF-kappa B.


Subject(s)
NF-kappa B/physiology , Receptors, Cell Surface/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Base Sequence , HIV Long Terminal Repeat , Humans , Mice , Molecular Sequence Data , Receptors, Cell Surface/drug effects , Receptors, Tumor Necrosis Factor
SELECTION OF CITATIONS
SEARCH DETAIL
...