Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Biosystems ; 108(1-3): 45-51, 2012.
Article in English | MEDLINE | ID: mdl-22289626

ABSTRACT

Apoptosis is an important physiological process which enables organisms to remove unwanted or damaged cells. A mathematical model of the extrinsic pro-apoptotic signaling pathway has been introduced by Eissing et al. (2007) and a bistable behavior with a stable death state and a stable life state of the reaction system has been established. In this paper, we consider a spatial extension of the extrinsic pro-apoptotic signaling pathway incorporating diffusion terms and make a model-based, numerical analysis of the apoptotic switch in the spatial dimension. For the parameter regimes under consideration it turns out that for this model diffusion homogenizes rapidly the concentrations which afterward are governed by the original reaction system. The activation of effector-caspase 3 depends on the space averaged initial concentration of pro-caspase 8 and pro-caspase 3 at the beginning of the process.


Subject(s)
Apoptosis/physiology , Caspase 3/metabolism , Caspase 8/metabolism , Models, Biological , Apraxia, Ideomotor , Feedback, Physiological , Mathematical Concepts , Signal Transduction , Systems Biology
2.
Oncogene ; 28(15): 1769-81, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19287455

ABSTRACT

It has been shown that tumor necrosis factor receptor-2 (TNFR2) stimulation leads to degradation of TNF receptor associated factor-2 (TRAF2) and inhibition of TNFR1-induced activation of NFkappaB and JNK. Here, we show that TRAF1 inhibits TNFR2-induced proteasomal degradation of TRAF2 and relieves TNFR1-induced activation of NFkappaB from the inhibitory effect of TNFR2. TRAF1 co-recruited with TRAF2 to both TNF receptors. Despite lacking an amino-terminal RING/zinc-finger domain, TRAF1 did not interfere with TNFR1-induced activation of JNK and NFkappaB. It is noted that physiological expression levels of TRAF1 enhanced NFkappaB activation and interleukin-8 (IL8) production induced by TNFR2. Thus, TRAF1 shifts the quality of integrated TNFR1-TNFR2 signaling from apoptosis induction to proinflammatory NFkappaB signaling.


Subject(s)
Receptors, Tumor Necrosis Factor, Type II/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Signal Transduction/physiology , TNF Receptor-Associated Factor 1/physiology , Cell Line, Tumor , Humans , Interleukin-8/biosynthesis , NF-kappa B/metabolism , TNF Receptor-Associated Factor 2/physiology
3.
Prog Mol Subcell Biol ; 34: 47-72, 2004.
Article in English | MEDLINE | ID: mdl-14979664

ABSTRACT

A central event in innate immunity is the activation of the NF-kappaB signaling pathway and up-regulation of NF-kappaB-dependent defense genes. Attack of mammals as well as of insects by microorganisms leads, among other things, to the activation of receptors of the Toll-like receptor group. Various adaptor proteins involving members of the TNF receptor-associated factor (TRAF) family channel these receptor-generated signals to conserved intracellular kinase cascades that finally lead to the activation of NF-kappaB and JNK. In vertebrates, TRAF proteins link these pathways also to IL-1R-related molecules and members of the TNF receptor superfamily, which orchestrate a variety of immunoregulatory processes of the innate but also of the adaptive immune system. In this review, we will focus on the similarities but also the differences in TRAF-dependent signaling pathways of mammals and insects.


Subject(s)
Drosophila/immunology , Mammals/immunology , Receptors, Tumor Necrosis Factor/physiology , Animals , Caenorhabditis elegans/immunology , Drosophila/genetics , Drosophila Proteins/immunology , Immunity, Innate , Membrane Glycoproteins/immunology , Models, Biological , Mutation , NF-kappa B/immunology , Receptors, Cell Surface/immunology , Signal Transduction , Toll-Like Receptor 5 , Toll-Like Receptors
4.
Cell Death Differ ; 10(1): 45-65, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12655295

ABSTRACT

A single mouse click on the topic tumor necrosis factor (TNF) in PubMed reveals about 50,000 articles providing one or the other information about this pleiotropic cytokine or its relatives. This demonstrates the enormous scientific and clinical interest in elucidating the biology of a molecule (or rather a large family of molecules), which began now almost 30 years ago with the description of a cytokine able to exert antitumoral effects in mouse models. Although our understanding of the multiple functions of TNF in vivo and of the respective underlying mechanisms at a cellular and molecular level has made enormous progress since then, new aspects are steadily uncovered and it appears that still much needs to be learned before we can conclude that we have a full comprehension of TNF biology. This review shortly covers some general aspects of this fascinating molecule and then concentrates on the molecular mechanisms of TNF signal transduction. In particular, the multiple facets of crosstalk between the various signalling pathways engaged by TNF will be addressed.


Subject(s)
Apoptosis/physiology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Proteins/metabolism , TNF Receptor-Associated Factor 1 , TNF Receptor-Associated Factor 2
5.
Apoptosis ; 7(5): 449-59, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12207178

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL/APO-2L) is a typical member of the TNF ligand family that induces apoptosis by activating the death receptors TRAIL-R1 and TRAIL-R2. TRAIL has attracted great attention in recent years as a promising anti cancer reagent because recombinant soluble TRAIL derivatives induce apoptosis in a broad range of tumor cells but not or only rarely in non-transformed cells. In this review we will address the putative role of TRAIL in cancer treatment in the light of the emerging importance of TRAIL in tumor surveillance and discuss the molecular basis of the cooperation of TRAIL and chemotherapeutic drugs. In particular, we debate controversial data in the literature concerning the cytotoxicity of different TRAIL derivatives on primary human cells.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Immunologic Surveillance/drug effects , Membrane Glycoproteins/metabolism , Neoplasms/drug therapy , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis Regulatory Proteins , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Humans , Immunologic Surveillance/genetics , Membrane Glycoproteins/pharmacology , Membrane Glycoproteins/therapeutic use , Neoplasms/genetics , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/therapeutic use
6.
J Biol Chem ; 276(47): 43708-12, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11557763

ABSTRACT

We show that tumor necrosis factor (TNF) and phorbol 12-myristate 13-acetate (PMA) induce TNF-related apoptosis-inducing ligand (TRAIL) in T cells. In cells deficient for NF-kappaB essential modulator (NEMO)/IKKgamma, an essential component of the NF-kappaB-inducing I-kappaB kinase (IKK) complex, induction of TRAIL expression was completely abrogated but was recovered in cells restored for IKKgamma expression. In cells deficient for receptor-interacting protein expression TNF, but not PMA-induced TRAIL expression was blocked. Inhibition of protein synthesis with cycloheximide blocked PMA, but not TNF-induced up-regulation of TRAIL. As both TNF and PMA rapidly induce NF-kappaB activation this suggests that NEMO/IKKgamma-dependent activation of the NF-kappaB pathway is necessary but not sufficient for up-regulation of TRAIL in T cells. The capability of the NF-kappaB pathway to induce the potent death ligand TRAIL may explain the reported proapoptotic features of this typically antiapoptotic pathway.


Subject(s)
Membrane Glycoproteins/biosynthesis , Protein Serine-Threonine Kinases/physiology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Base Sequence , DNA Primers , Humans , I-kappa B Kinase , Jurkat Cells , NF-kappa B/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Up-Regulation/drug effects
7.
Oncogene ; 20(30): 4101-6, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11494138

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL) is a typical member of the tumor necrosis factor (TNF) ligand family that is expressed as a type II membrane protein (memTRAIL) and signals apoptosis via the death domain-containing receptors TRAIL-R1 and -2. Soluble recombinant derivatives of TRAIL (sTRAIL) are considered as novel tumors therapeutics because of their selective apoptosis inducing activity in a variety of human tumors but not in normal cells. Using antagonistic antigen-binding fragment (Fab) preparations of TRAIL-R1- and TRAIL-R2-specific antibodies, we demonstrate in this study that TRAIL-R1 becomes activated by both the soluble and the membrane-bound form of the ligand, whereas TRAIL-R2 becomes only activated by memTRAIL or soluble TRAIL secondarily cross-linked by antibodies. Furthermore, we show that the restricted signal capacity of sTRAIL can be readily converted into a fully signal competent memTRAIL-like molecule, i.e. a TRAIL-R2 stimulating ligand, by genetic fusion to an antibody derivative that allows antigen-dependent 'immobilization' of the fusion protein to cell surfaces. We conclude that antibody targeting-dependent activation can be used to design selective therapeutics derived of those ligands of the TNF family that are biologically inactive in their soluble form.


Subject(s)
Antigens, Surface/immunology , Membrane Glycoproteins/pharmacology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibody Specificity , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Apoptosis Regulatory Proteins , COS Cells , Chlorocebus aethiops , Drug Design , HeLa Cells/drug effects , Humans , Immunoglobulin Fab Fragments , Jurkat Cells/drug effects , KB Cells/drug effects , Ligands , Membrane Glycoproteins/chemistry , Membrane Proteins/chemistry , Membrane Proteins/pharmacology , Neoplasm Proteins/drug effects , Neoplasm Proteins/immunology , Neoplasm Proteins/physiology , Protein Structure, Tertiary , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Signal Transduction/physiology , Solubility , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/chemistry
8.
Cell Signal ; 13(6): 389-400, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11384837

ABSTRACT

The TNF-receptor-associated factor (TRAF) family is a phylogenetically conserved group of scaffold proteins that link receptors of the IL-1R/Toll and TNF receptor family to signalling cascades, leading to the activation of NF-kappaB and mitogen-activated protein kinases. Furthermore, TRAF proteins serve as a docking platform for a variety of regulators of these signalling pathways and are themselves often regulated at the transcriptional and posttranslational level. In this review, we address the structural and molecular basis of TRAF protein functions and highlight their role in cytokine signalling.


Subject(s)
Multigene Family , Protein Serine-Threonine Kinases , Receptors, Cytokine/metabolism , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction , Animals , Carrier Proteins/metabolism , Enzyme Activation , Humans , Kinetics , MAP Kinase Signaling System , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , NF-kappa B/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor/genetics , TNF Receptor-Associated Factor 1 , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism
9.
J Biol Chem ; 276(35): 32585-90, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11384965

ABSTRACT

Fas, a death domain-containing member of the tumor necrosis factor receptor family and its ligand FasL have been predominantly studied with respect to their capability to induce cell death. However, a few studies indicate a proliferation-inducing signaling activity of these molecules too. We describe here a novel signaling pathway of FasL and the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) that triggers transcriptional activation of the proto-oncogene c-fos, a typical target gene of mitogenic pathways. FasL- and TRAIL-mediated up-regulation of c-Fos was completely dependent on the presence of Fas-associated death domain protein (FADD) and caspase-8, but caspase activity seemed to be dispensable as a pan inhibitor of caspases had no inhibitory effect. Upon overexpression of the long splice form of cellular FADD-like interleukin-1-converting enzyme (FLICE) inhibitory protein (cFLIP) in Jurkat cells, FasL- and TRAIL-induced up-regulation of c-Fos was almost completely blocked. The short splice form of FLIP, however, showed a rather stimulatory effect on c-Fos induction. Together these data demonstrate the existence of a death receptor-induced, FADD- and caspase-8-dependent pathway leading to c-Fos induction that is inhibited by the long splice form FLIP-L.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Caspases/metabolism , Gene Expression Regulation , Genes, fos , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/physiology , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology , Alternative Splicing , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/genetics , Caspase 8 , Caspase 9 , Fas Ligand Protein , Fas-Associated Death Domain Protein , Humans , Jurkat Cells , Models, Biological , Proto-Oncogene Mas , Proto-Oncogene Proteins c-fos/genetics , Recombinant Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transfection
10.
Mol Cell Biol ; 21(12): 3964-73, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11359904

ABSTRACT

The caspase 8 homologue FLICE-inhibitory protein (cFLIP) is a potent negative regulator of death receptor-induced apoptosis. We found that cFLIP can be upregulated in some cell lines under critical involvement of the NF-kappaB pathway, but NF-kappaB activation was clearly not sufficient for cFLIP induction in all cell lines. Treatment of SV80 cells with the proteasome inhibitor N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG-132) or geldanamycin, a drug interfering with tumor necrosis factor (TNF)-induced NF-kappaB activation, inhibited TNF-induced upregulation of cFLIP. Overexpression of a nondegradable IkappaBalpha mutant (IkappaBalpha-SR) or lack of IkappaB kinase gamma expression completely prevented phorbol myristate acetate-induced upregulation of cFLIP mRNA in Jurkat cells. These data point to an important role for NF-kappaB in the regulation of the cFLIP gene. SV80 cells normally show resistance to TNF-related apoptosis-inducing ligand (TRAIL) and TNF, as apoptosis can be induced only in the presence of low concentrations of cycloheximide (CHX). However, overexpression of IkappaBalpha-SR rendered SV80 cells sensitive to TRAIL-induced apoptosis in the absence of CHX, and cFLIP expression was able to reverse the proapoptotic effect of NF-kappaB inhibition. Western blot analysis further revealed that cFLIP, but not TRAF1, A20, and cIAP2, expression levels rapidly decrease upon CHX treatment. In conclusion, these data suggest a key role for cFLIP in the antiapoptotic response of NF-kappaB activation.


Subject(s)
Carrier Proteins/physiology , I-kappa B Proteins , Intracellular Signaling Peptides and Proteins , NF-kappa B/metabolism , Antibodies, Monoclonal/pharmacology , Apoptosis , Benzoquinones , CASP8 and FADD-Like Apoptosis Regulating Protein , CD40 Antigens/immunology , Carrier Proteins/genetics , Cell Line , Cycloheximide/pharmacology , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Interleukin-1/pharmacology , Jurkat Cells , Lactams, Macrocyclic , Leupeptins/pharmacology , Mutation , NF-KappaB Inhibitor alpha , Quinones/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
11.
Oncogene ; 20(5): 571-80, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11313989

ABSTRACT

One of the main functions of the tumor suppressor p53 is the induction of programmed cell death. Here we investigated in detail the molecular mechanisms that underlay p53 transactivation-dependent apoptosis in the human colon cancer cell line DLD-1. Although p53 upregulated the death receptors Fas, TRAIL-R1 and TRAIL-R2 in this cell line, p53-induced cell death occurred without detectable caspase-8 activation whereas, activation of caspase-9 and caspase-3 was readily observed. In addition to the upregulation of death receptors, p53 induced the pro-apoptotic Bcl-2 family members Bik and Bak and downregulated the anti-apoptotic Bcl-xL protein. Moreover, in RNase protection assay analyses as well as in reporter gene analyses we found a p53-dependent upregulation of the death receptor-inhibitory protein cFLIP. Together, these data argue for a p53-mediated activation of the mitochondrial pathway of apoptosis. In contrast to recently published data obtained in different cellular systems, there was no evidence for an essential role of NF-kappaB in p53-induced cell death. Moreover, induction of p53 interfered with TNF-induced NF-kappaB activation independently from apoptosis-induction.


Subject(s)
Apoptosis/physiology , Carrier Proteins/genetics , Caspases/physiology , Intracellular Signaling Peptides and Proteins , NF-kappa B/physiology , Tumor Suppressor Protein p53/physiology , Base Sequence , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/biosynthesis , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Enzyme Activation , Gene Expression Regulation , Humans , Molecular Sequence Data , NF-kappa B/genetics , Transcriptional Activation/physiology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Up-Regulation
12.
Int J Biochem Cell Biol ; 33(1): 19-32, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11167129

ABSTRACT

Tumor necrosis factor (TNF) is the prototypic member of the TNF ligand family and has a key role in the regulation of inflammatory processes. TNF exerts its functions by interaction with the death domain-containing TNF-receptor 1 (TNF-R1) and the non-death domain-containing TNF-receptor 2 (TNF-R2), both members of a receptor family complementary to the TNF ligand family. Due to the prototypic features of the TNF receptors and their importance for the regulation of inflammation, the signal transduction mechanisms utilized by these receptors have been extensively studied. Several proteins that interact directly or indirectly with the cytoplasmic domains of TNF-R1 and TNF-R2 have been identified in the recent years giving ideas how these receptors are connected to the apoptotic pathway and the signaling cascades leading to activation of NF-kappaB and JNK. Of special interest are TNF receptor-associated factor (TRAF) 1 and 2, which defines a novel group of adaptor proteins involved in signal transduction by most members of the TNF receptor family, of IL-1 receptor and IL-17 receptor as well as some members of the TOLL-like receptor family. TRAF 2 is currently the best-characterized TRAF family member, having a key role in mediating TNF-R1-induced activation of NF-kappaB and JNK. Moreover, recent studies suggest that TRAF 2 represents an integration point for pro- and antiapoptotic signals. This review focuses on the molecular mechanisms that underlay signal initiation by TNF-R1 and TNF-R2, with particular consideration of the role of TRAF 2, and highlights the importance of this molecule for the integration of such antagonizing pathways as death induction and NF-kappaB-mediated surviving signals.


Subject(s)
Proteins/metabolism , Proteins/physiology , Animals , Cell Death , Cell Survival , Humans , Ligands , Models, Biological , Mutation , NF-kappa B/metabolism , Proteins/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , TNF Receptor-Associated Factor 1 , TNF Receptor-Associated Factor 2
13.
Blood ; 97(5): 1321-9, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222376

ABSTRACT

Vascular endothelial growth factor (VEGF) induces both angiogenesis and an increase in vascular permeability, 2 processes that are considered to be important for both tumor growth and the delivery of drugs to the site of tumors. This study demonstrates that transmembrane expression of tumor necrosis factor (tmTNF) is up-regulated in the endothelium of a murine methylcholanthrene (meth A)-induced sarcoma in comparison to the adjacent normal dermal vasculature and is also present on cultivated human endothelial cells. It is further shown that tmTNF is required for VEGF-mediated endothelial hyperpermeability in vitro and in vivo. This permissive activity of TNF appears to be selective, because anti-TNF antibodies ablated the VEGF-induced permeability but not proliferation of cultivated human endothelial cells. Furthermore, tnf gene-deficient mice show no obvious defects in vascularization and develop normally but failed to respond to administration of VEGF with an increase in vascular permeability. Subsequent studies indicated that the tmTNF and VEGF signaling pathways converge at the level of a secondary messenger, the "stress-activated protein kinase-2" (SAPK-2)/p38: (1) up-regulated endothelial expression of tmTNF resulted in the continuous activation of SAPK-2/p38 in vitro, and (2) an inhibitor of SAPK-2/p38 activation abolished the vascular permeability activity of VEGF in vivo. In conclusion, the study's finding that continuous autocrine signaling by tmTNF sensitizes endothelial cells to respond to VEGF by increasing their vascular permeability provides new therapeutic concepts for manipulating vascular hyperpermeability.


Subject(s)
Capillary Permeability/drug effects , Endothelial Growth Factors/pharmacology , Lymphokines/pharmacology , Tumor Necrosis Factor-alpha/physiology , Animals , Autocrine Communication/drug effects , Dipeptides/pharmacology , Endothelial Growth Factors/metabolism , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Humans , Hydroxamic Acids/pharmacology , Immunohistochemistry , Lymphokines/metabolism , Membrane Glycoproteins/pharmacology , Membrane Glycoproteins/physiology , Methylcholanthrene , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/pharmacology , Neoplasm Proteins/physiology , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/chemically induced , Thromboplastin/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Cord/cytology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
J Immunol ; 164(12): 6193-8, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843670

ABSTRACT

We have previously reported that the CD14+ monocytic subpopulation of human PBMC induces programmed cell death (apoptosis) in cocultured endothelial cells (EC) when stimulated by bacterial endotoxin (LPS). Apoptosis is mediated by two routes, first via transmembrane TNF-alpha (mTNF) expressed on PBMC and, in addition, by TNF-independent soluble factors that trigger apoptosis in EC. Neutralizing anti-TNF mAb completely blocked coculture-mediated apoptosis, despite the fact that there should have been additional soluble cell death factors. This led to the hypothesis that a reverse signal is transmitted from the TNF receptor on EC to monocytes (MO) via mTNF that prevents the production of soluble apoptotic factors. Here we have tested this hypothesis. The results support the idea of a bidirectional cross-talk between MO and EC. Peripheral blood MO, MO-derived macrophages (MPhi), or the monocytic cell line Mono Mac 6 were preincubated with human microvascular EC that constitutively express TNF receptor type I (TNF-R1) and subsequently stimulated with LPS. Cell-free supernatants of these preparations no longer induced EC apoptosis. The preincubation of MO/MPhi with TNF-reactive agents, such as mAb and soluble receptors, also blocked the production of death factors, providing further evidence for reverse signaling via mTNF. Finally, we show that reverse signaling through mTNF mediated LPS resistance in MO/MPhi as indicated by the down-regulation of LPS-induced soluble TNF and IL-6 as well as IL-1 and IL-10.


Subject(s)
Lipopolysaccharides/immunology , Macrophages/immunology , Membrane Proteins/immunology , Monocytes/immunology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/blood , Antigens, CD/physiology , Apoptosis/immunology , Cell Death/immunology , Cell Line , Cell-Free System/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Humans , Immunity, Innate , Immunoglobulin Fab Fragments/pharmacology , Interleukin-1/antagonists & inhibitors , Interleukin-1/metabolism , Interleukin-10/antagonists & inhibitors , Interleukin-10/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Macrophages/metabolism , Monocytes/metabolism , Receptors, Tumor Necrosis Factor/blood , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Solubility , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
15.
J Biol Chem ; 275(32): 24357-66, 2000 Aug 11.
Article in English | MEDLINE | ID: mdl-10823821

ABSTRACT

In HeLa cells, induction of apoptosis and nuclear factor kappaB (NF-kappaB) activation initiated by TRAIL/Apo2L or the agonistic Apo1/Fas-specific monoclonal antibody anti-APO-1 require the presence of cycloheximide (CHX). Inhibition of caspases prevented TRAIL/anti-APO-1-induced apoptosis, but not NF-kappaB activation, indicating that both pathways bifurcate upstream of the receptor-proximal caspase-8. Under these conditions, TRAIL and anti-APO-1 up-regulated the expression of the known NF-kappaB targets interleukin-6, cellular inhibitor of apoptosis 2 (cIAP2), and TRAF1 (TRAF, tumor necrosis factor receptor-associate factor). In the presence of CHX, the stable overexpression of a deletion mutant of the Fas-associated death domain molecule FADD comprising solely the death domain of the molecule but lacking its death effector domain (FADD-(80-208)) led to the same response pattern as TRAIL or anti-APO-1 treatment. Moreover, the ability of death receptors to induce NF-kappaB activation was drastically reduced in a FADD-deficient Jurkat cell line. TRAIL-, anti-APO-1-, and FADD-(80-208)-initiated gene induction was blocked by a dominant-negative mutant of TRAF2 or the p38 kinase inhibitor SB203580, similar to tumor necrosis factor receptor-1-induced NF-kappaB activation. CHX treatment rapidly down-regulated endogenous cFLIP protein levels, and overexpression of cellular FLICE inhibitory protein (cFLIP) inhibited death receptor-induced NF-kappaB activation. Thus, a novel functional role of cFLIP as a negative regulator of gene induction by death receptors became apparent.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Gene Expression Regulation/physiology , NF-kappa B/metabolism , Proteins/metabolism , Receptors, Tumor Necrosis Factor/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Antibodies, Monoclonal/pharmacology , Apoptosis , Carrier Proteins/chemistry , Carrier Proteins/genetics , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Line , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Fas-Associated Death Domain Protein , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Kinetics , Receptors, TNF-Related Apoptosis-Inducing Ligand , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , TNF Receptor-Associated Factor 1 , Transcriptional Activation , fas Receptor/physiology
16.
J Biol Chem ; 275(41): 32208-13, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-10807904

ABSTRACT

Overexpression of the tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors, TRAIL-R1 and TRAIL-R2, induces apoptosis and activation of NF-kappaB in cultured cells. In this study, we have demonstrated differential signaling capacities by both receptors using either epitope-tagged soluble TRAIL (sTRAIL) or sTRAIL that was cross-linked with a monoclonal antibody. Interestingly, sTRAIL was sufficient for induction of apoptosis only in cell lines that were killed by agonistic TRAIL-R1- and TRAIL-R2-specific IgG preparations. Moreover, in these cell lines interleukin-6 secretion and NF-kappaB activation were induced by cross-linked or non-cross-linked anti-TRAIL, as well as by both receptor-specific IgGs. However, cross-linking of sTRAIL was required for induction of apoptosis in cell lines that only responded to the agonistic anti-TRAIL-R2-IgG. Interestingly, activation of c-Jun N-terminal kinase (JNK) was only observed in response to either cross-linked sTRAIL or anti-TRAIL-R2-IgG even in cell lines where both receptors were capable of signaling apoptosis and NF-kappaB activation. Taken together, our data suggest that TRAIL-R1 responds to either cross-linked or non-cross-linked sTRAIL which signals NF-kappaB activation and apoptosis, whereas TRAIL-R2 signals NF-kappaB activation, apoptosis, and JNK activation only in response to cross-linked TRAIL.


Subject(s)
Apoptosis , Mitogen-Activated Protein Kinases/metabolism , Receptor Aggregation , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Antibodies, Monoclonal/immunology , Apoptosis Regulatory Proteins , Enzyme Activation , Flow Cytometry , Humans , Immunoglobulin G/immunology , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases , Membrane Glycoproteins/agonists , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , NF-kappa B/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/agonists , Staphylococcal Protein A/immunology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
17.
EMBO J ; 18(11): 3034-43, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10357816

ABSTRACT

Several members of the tumour necrosis factor receptor (TNF-R) superfamily can induce cell death. For TNF-R1, Fas/APO-1, DR3, DR6, TRAIL-R1 and TRAIL-R2, a conserved 'death domain' in the intracellular region couples these receptors to activation of caspases. However, it is not yet known how TNF receptor family members lacking a death domain, such as TNF-R2, CD40, LT-betaR, CD27 or CD30, execute their death-inducing capability. Here we demonstrate in different cellular systems that cytotoxic effects induced by TNF-R2, CD40 and CD30 are mediated by endogenous production of TNF and autotropic or paratropic activation of TNF-R1. In addition, stimulation of TNF-R2 and CD40 synergistically enhances TNF-R1-induced cytotoxicity. These findings describe a novel pro-apoptotic mechanism induced by some members of the TNF-R family.


Subject(s)
Antigens, CD/physiology , Apoptosis , CD40 Antigens/physiology , Ki-1 Antigen/physiology , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies/pharmacology , Antigens, CD/chemistry , Antigens, CD/genetics , Apoptosis/drug effects , CD40 Antigens/genetics , Caspase Inhibitors , Caspases/metabolism , Fas Ligand Protein , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/pharmacology , Membrane Glycoproteins/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/physiology , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Signal Transduction , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Up-Regulation , bcl-X Protein , Interferon gamma Receptor
18.
Cytokine Growth Factor Rev ; 10(1): 15-26, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10379909

ABSTRACT

Just four years ago the first two members of a new family of molecules involved in signal transduction by members of the TNF receptor superfamily were described and designated TNF Receptor Associated Factors (TRAFs). In the meantime six human and murine TRAFs as well as a TRAF protein from C. elegans have been molecularly cloned. From our current point of view, TRAF proteins appear to represent multifunctional signal adaptors, tightly embedded in a network of signals culminating in the activation of kinase cascades that finally lead to the activation of c-Jun N-terminal kinase. p38 mitogen activated protein kinase, and the transcription factor NF-kappaB, thereby also affecting the balance between survival and cell death. Some of the activities of the individual TRAF family members may be redundant although transgenic knockout animal models have already shown that crucial signaling pathways for single TRAF molecules in vivo can be defined.


Subject(s)
Cytokines/physiology , Multigene Family , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Humans , Molecular Sequence Data , Receptors, Tumor Necrosis Factor/genetics , Sequence Homology, Amino Acid , Viral Matrix Proteins
19.
J Biol Chem ; 274(27): 19368-74, 1999 Jul 02.
Article in English | MEDLINE | ID: mdl-10383449

ABSTRACT

To understand how the TNF receptor-associated factor 1 (TRAF1) is transcriptionally regulated, in vitro DNA binding assays, promoter-reporter gene assays, and RNase protection assays were performed with the human TRAF1 gene. Binding of NF-kappaB to three of five putative binding sites within the human TRAF1 promoter was found in electrophoretic mobility shift assay studies, and analysis of TRAF1 gene promoter luciferase constructs confirmed the functional importance of these elements. Moreover, triggering of TNF-R1, CD40, and the interleukin-1 receptor resulted in transcription of the TRAF1 gene, whereas receptors that are not activators or only poor activators of NF-kappaB in HeLa cells failed to show a significant TRAF1 induction. Because it has been shown that members of the TRAF family are involved in activation of NF-kappaB and the c-Jun N-terminal kinase (JNK) by the interleukin-1 receptor and members of the TNF receptor superfamily, a role of TRAF1 in receptor cross-talk and/or feedback regulation of activated receptor signaling complexes can be suggested. In fact, we found that TNF-induced activation of JNK is prolonged in transfectants overexpressing TRAF1, whereas overexpression of a deletion mutant of TRAF1 in which the N-terminal part had been replaced by the green fluorescent protein interfered with TNF-induced activation of NF-kappaB and JNK.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , NF-kappa B/metabolism , Proteins/genetics , Receptors, Tumor Necrosis Factor/genetics , Up-Regulation , Base Sequence , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , Molecular Sequence Data , Mutagenesis , Promoter Regions, Genetic , Sequence Deletion , TNF Receptor-Associated Factor 1 , Transcription, Genetic
20.
Eur J Immunol ; 29(6): 1785-92, 1999 06.
Article in English | MEDLINE | ID: mdl-10382740

ABSTRACT

TWEAK is a recently cloned novel member of the TNF ligand family. Here we show that soluble TWEAK is sufficient to induce apoptosis in Kym-1 cells within 18 h. TWEAK-induced apoptosis is indirect and is mediated by the interaction of endogenous TNF and TNF receptor (TNFR)1, as each TNFR1-Fc, neutralizing TNF-specific antibodies and TNFR1-specific Fab fragments efficiently antagonize cell death induction. In addition to this indirect mode of action, co-stimulation of Kym-1 cells with TWEAK enhances TNFR1-mediated cell death induction. In contrast to TNF, TWEAK does only modestly activate NF-kappaB or c-jun N-terminal kinase (JNK) in Kym-1 cells. Although TWEAK binding to Kym-1 cells is easily detectable by flow cytometric analysis, we found neither evidence for expression of the recently identified TWEAK receptor Apo3/TRAMP/wsl/DR3/LARD, nor indications for direct interactions of TWEAK with TNFR. Together, these characteristics of TWEAK-induced signaling in Kym-1 cells argue for the existence of an additional, still undefined non-death domain-containing TWEAK receptor in Kym-1 cells.


Subject(s)
Antigens, CD/physiology , Apoptosis/physiology , Carrier Proteins/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Base Sequence , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cell Line , Cytokine TWEAK , Humans , NF-kappa B/metabolism , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , Receptors, Tumor Necrosis Factor, Member 25 , Receptors, Tumor Necrosis Factor, Type I , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Tumor Necrosis Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...