Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Appl Microbiol Biotechnol ; 98(11): 4839-51, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24728757

ABSTRACT

Molecular studies have added significantly to understanding of the role of fungi and fungal enzymes in the efficient biomass conversion, which takes place in the fungus garden of leaf-cutting ants. It is now clear that the fungal symbiont expresses the full spectrum of genes for degrading cellulose and other plant cell wall polysaccharides. Since the start of the genomics era, numerous interesting studies have especially focused on evolutionary, molecular, and organismal aspects of the biological and biochemical functions of the symbiosis between leaf-cutting ants (Atta spp. and Acromyrmex spp.) and their fungal symbiont Leucoagaricus gongylophorus. Macroscopic observations of the fungus-farming ant colony inherently depict the ants as the leading part of the symbiosis (the myrmicocentric approach, overshadowing the mycocentric aspects). However, at the molecular level, it is fungal enzymes that enable the ants to access the nutrition embedded in recalcitrant plant biomass. Our hypothesis is that the evolutionary events that established fungus-farming practice were predisposed by a fascinating fungal evolution toward increasing attractiveness to ants. This resulted in the ants allowing the fungus to grow in the nests and began to supply plant materials for more fungal growth. Molecular studies also confirm that specialized fungal structures, the gongylidia, with high levels of proteins and rich blend of enzymes, are essential for symbiosis. Harvested and used as ant feed, the gongylidia are the key factor for sustaining the highly complex leaf-cutting ant colony. This microbial upgrade of fresh leaves to protein-enriched animal feed can serve as inspiration for modern biorefinery technology.


Subject(s)
Agaricales/physiology , Ants/microbiology , Ants/physiology , Polysaccharides/metabolism , Symbiosis , Agaricales/metabolism , Animals
2.
Philos Trans A Math Phys Eng Sci ; 368(1927): 4313-30, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20732889

ABSTRACT

The use of the water-oil interface provides significant advantages in the synthesis of inorganic nanostructures. Employing the water-toluene interface, luminescent CdS nanocrystals have been obtained at a relatively modest temperature of 35 degrees C. The diameters of the particulates can be varied between 1.0 and 5.0 nm. In addition, we have devised a new method for transferring thin films at the water-toluene interface onto solid substrates. Using this method, thin films consisting of Au and Ag nanocrystals spread over very large areas (square centimetres) are obtained in a single step. These films are directly usable as ingredients of functional devices. We show this by constructing a working amine sensor based on films of Au nanocrystals. The materials obtained have been characterized by X-ray diffraction, scanning and transmission electron microscopy, absorption and emission spectroscopy and charge transport measurements.

3.
Toxicol In Vitro ; 23(8): 1541-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19422909

ABSTRACT

The organotin compounds trimethyltin (TMT), triethyltin (TET) and tributyltin (TBT) show different organotoxicities in vivo. While TMT and TET induce a strong neurotoxicity accompanied by microglial and astroglial activation, TBT rather effects the immune system. Previously, we have shown in an in vitro co-culture model that microglial cells can be activated by TMT in the presence of astrocytes. In this study, we wanted to investigate (a) if the neurotoxic organotin compound TET can also activate microglial cells in vitro similar to TMT and (b) if differences between the neurotoxicants TMT and TET on the one side and TBT on the other exist concerning microglial activation. Therefore, purified microglial and astroglial cell cultures from neonatal rat brains were treated either alone or in co-cultures for 24h with different concentrations of TMT, TET or TBT and the basal cytotoxicity and nitric oxide formation was determined. Furthermore, morphological changes of astrocytes were examined. Our results show that microglial activation can be increased in subcytolethal concentrations, but only in the presence of astrocytes and not in microglial cell cultures alone. This increase was induced by the neurotoxicants TMT and TET but not by TBT. Taken together, the differing microglia activating effect of the organotin compounds may contribute to the differing neurotoxic potential of this group of chemicals in vivo. In addition, our results emphasize the need for co-culture systems when studying interactions between different cell types for toxicity assessment.


Subject(s)
Astrocytes/drug effects , Microglia/drug effects , Trialkyltin Compounds/toxicity , Triethyltin Compounds/toxicity , Trimethyltin Compounds/toxicity , Animals , Cells, Cultured , Coculture Techniques , Dose-Response Relationship, Drug , Nitric Oxide/biosynthesis , Rats , Rats, Wistar
4.
Oncogene ; 26(39): 5722-32, 2007 Aug 23.
Article in English | MEDLINE | ID: mdl-17369858

ABSTRACT

Tumor necrosis factor (TNF) is a pro-inflammatory cytokine exerting pleiotropic effects on endothelial cells. Depending on the vascular context it can induce endothelial cell activation and survival or death. The microenvironmental cues determining whether endothelial cells will survive or die, however, have remained elusive. Here we report that integrin ligation acts permissive for TNF-induced protein kinase B (PKB/Akt) but not nuclear factor (NF)-kappaB activation. Concomitant activation of PKB/Akt and NF-kappaB is essential for the survival of endothelial cells exposed to TNF. Active PKB/Akt strengthens integrin-dependent endothelial cell adhesion, whereas disruption of actin stress fibers abolishes the protective effect of PKB/Akt. Integrin-mediated adhesion also represses TNF-induced JNK activation, but JNK activity is not required for cell death. The alphaVbeta3/alphaVbeta5 integrin inhibitor EMD121974 sensitizes endothelial cells to TNF-dependent cytotoxicity and active PKB/Akt attenuates this effect. Interferon gamma synergistically enhanced TNF-induced endothelial cell death in all conditions tested. Taken together, these observations reveal a novel permissive role for integrins in TNF-induced PKB/Akt activation and prevention of TNF-induced death distinct of NF-kappaB, and implicate the actin cytoskeleton in PKB/Akt-mediated cell survival. The sensitizing effect of EMD121974 on TNF cytotoxicity may open new perspectives to the therapeutic use of TNF as anticancer agent.


Subject(s)
Apoptosis/physiology , Cell Adhesion , Endothelium, Vascular/cytology , Integrin alphaVbeta3/metabolism , Integrins/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Vitronectin/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Actins/metabolism , Blotting, Western , Cells, Cultured , Cytoskeleton/metabolism , Electrophoretic Mobility Shift Assay , Endothelium, Vascular/metabolism , Flow Cytometry , Humans , In Situ Nick-End Labeling , Integrin alphaVbeta3/antagonists & inhibitors , Integrins/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , NF-kappa B/genetics , Phosphorylation , Receptors, Vitronectin/antagonists & inhibitors , Signal Transduction , Spheroids, Cellular
5.
Mol Genet Genomics ; 269(5): 699-705, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12838413

ABSTRACT

A linear plasmid is widespread among isolates of the obligate biotrophic fungus Blumeria graminis f.sp. hordei (synonym Erysiphe graminis) (Bgh), the organism that causes the disease powdery mildew on barley. We cloned and sequenced the entire plasmid of 7965 bp. The plasmid contains two identical terminal inverted repeats (TIR) of 610 bp. Two ORFs are present on opposite strands, one encoding a phage-type DNA polymerase and the other a phage-type RNA polymerase. Two large transcripts of approximately 4.2 and 5.6 kb were identified in conidia, germinating conidia and Bgh -infected barley leaves, indicating that the polymerases are transcribed at most stages of the lifecycle. The transcription start sites were localised within the TIR regions, where a putative 11-bp ARS consensus sequence was also identified. To follow the sexual transmission of the plasmid we screened 27 Bgh isolates for mitochondrial polymorphisms. One polymorphism allowed us to carry out a cross between two isolates that differed in both mitochondrial genotype and presence/absence of the Bgh plasmid. The plasmid was transmitted independently of the origin of the mitochondria. No transfer of the plasmid was observed between two Bgh isolates that were co-cultivated for 1.5 years on a common susceptible barley variety. The plasmid appears to be an autonomous replicon with no phenotypic effect on Bgh.


Subject(s)
Ascomycota/genetics , Plasmids , Base Sequence , DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed RNA Polymerases/genetics , Gene Expression , Phylogeny , Transcription, Genetic
6.
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
7.
Nature ; 408(6809): 211-6, 2000 Nov 09.
Article in English | MEDLINE | ID: mdl-11089976

ABSTRACT

Cytokines are multifunctional mediators that classically modulate immune activity by receptor-mediated pathways. Macrophage migration inhibitory factor (MIF) is a cytokine that has a critical role in several inflammatory conditions but that also has endocrine and enzymatic functions. The molecular targets of MIF action have so far remained unclear. Here we show that MIF specifically interacts with an intracellular protein, Jab1, which is a coactivator of AP-1 transcription that also promotes degradation of the cyclin-dependent kinase inhibitor p27Kip1 (ref. 10). MIF colocalizes with Jab1 in the cytosol, and both endogenous and exogenously added MIF following endocytosis bind Jab1. MIF inhibits Jab1- and stimulus-enhanced AP-1 activity, but does not interfere with the induction of the transcription factor NFkappaB. Jab1 activates c-Jun amino-terminal kinase (JNK) activity and enhances endogenous phospho-c-Jun levels, and MIF inhibits these effects. MIF also antagonizes Jab1-dependent cell-cycle regulation by increasing p27Kip1 expression through stabilization of p27Kip1 protein. Consequently, Jab1-mediated rescue of fibroblasts from growth arrest is blocked by MIF. Amino acids 50-65 and Cys 60 of MIF are important for Jab1 binding and modulation. We conclude that MIF may act broadly to negatively regulate Jab1-controlled pathways and that the MIF-Jab1 interaction may provide a molecular basis for key activities of MIF.


Subject(s)
Cell Cycle Proteins , Cell Cycle/physiology , DNA-Binding Proteins/physiology , JNK Mitogen-Activated Protein Kinases , Macrophage Migration-Inhibitory Factors/physiology , Transcription Factor AP-1/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins , COP9 Signalosome Complex , Cell Line , Cyclin-Dependent Kinase Inhibitor p27 , Gene Expression Regulation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Luciferases/genetics , MAP Kinase Kinase 4 , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Peptide Hydrolases , Precipitin Tests , Protein Binding , Proto-Oncogene Proteins c-jun/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factor AP-1/antagonists & inhibitors
8.
Microsc Res Tech ; 50(3): 243-50, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10891890

ABSTRACT

Tumor necrosis factor (TNF) exists in two bioactive forms, the membrane integrated form and the proteolytically derived soluble cytokine. Both forms of TNF are involved in a variety of different physiological and pathophysiological situations. Here we analyzed different human and mouse TNF-specific reagents for their ability to determine the expression of membrane-expressed TNF. The data prove some antibodies to be very useful for the analysis of transmembrane TNF expression because these antibodies distinguish between the transmembrane form of TNF and soluble TNF bound to cellular TNF receptors. In addition, we found that recombinant human TNF receptor fusion proteins are advantageous tools to analyze both human and mouse transmembrane TNF expression.


Subject(s)
Cell Membrane/chemistry , Tumor Necrosis Factor-alpha/analysis , Animals , Antibodies, Monoclonal/immunology , CHO Cells , Cricetinae , Flow Cytometry , HeLa Cells , Humans , Mice , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
9.
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
10.
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
12.
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
13.
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
14.
Endocrinology ; 140(7): 3219-27, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385418

ABSTRACT

Tumor necrosis factor-alpha (TNFalpha) is a potential mediator of beta cell destruction in insulin-dependent diabetes mellitus. We have studied TNF-responsive pathways leading to apoptosis in beta cells. Primary beta cells express low levels of the type I TNF receptor (TNFR1) but do not express the type 2 receptor (TNFR2). Evidence for TNFR1 expression on beta cells came from flow cytometry using monoclonal antibodies specific for TNFR1 and TNFR2 and from RT-PCR of beta cell RNA. NIT-1 insulinoma cells similarly expressed TNFR1 (at higher levels than primary beta cells) as detected by flow cytometry and radio-binding studies. TNF induced NF-kappaB activation in both primary islet cells and NIT-1 cells. Apoptosis in response to TNFalpha was observed in NIT-1 cells whereas apoptosis of primary beta cells required both TNFalpha and interferon-gamma (IFNgamma). Apoptosis could be prevented in NIT-1 cells by expression of dominant negative Fas-associating protein with death domain (dnFADD). Apoptosis in NIT-1 cells was increased by coincubation with IFNgamma, which also increased caspase 1 expression. These data show that TNF-activated pathways capable of inducing apoptotic cell death are present in beta cells. Caspase activation is the dominant pathway of TNF-induced cell death in NIT-1 cells and may be an important mechanism of beta cell damage in insulin-dependent diabetes mellitus.


Subject(s)
Apoptosis/drug effects , Insulinoma/pathology , Islets of Langerhans/physiology , Pancreatic Neoplasms/pathology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/physiology , Caspase 1/metabolism , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Female , Insulinoma/metabolism , Insulinoma/physiopathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred NOD , NF-kappa B/physiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/physiopathology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Cells, Cultured/drug effects
15.
J Biol Chem ; 274(25): 18107-12, 1999 Jun 18.
Article in English | MEDLINE | ID: mdl-10364265

ABSTRACT

Tumor necrosis factor (TNF) exists in two bioactive forms, the membrane-integrated form and the proteolytically derived soluble cytokine. Cells that produce TNF are often responsive to TNF, allowing autocrine/juxtacrine feedback loops. However, whether the membrane form of TNF is involved in such regulatory circuits is unclear. Here we demonstrate that HeLa cells, expressing a permanently membrane-integrated mutant form of TNF, constitutively express TNF.TNF receptor complexes at their cell surface. These cells show a permanent activation of the transcription factor NF-kappaB, exert constitutive p38 mitogen-activated protein kinase activity, and produce high amounts of interleukin-6. In parallel, transmembrane TNF-expressing HeLa cells display high sensitivity to cycloheximide or interferon-gamma, similar to untransfected cells treated with these agents in combination with sTNF. Moreover, cycloheximide-induced apoptosis in transmembrane TNF transfectants can be blocked by the caspase inhibitor zVAD-fmk and does not necessarily need cell to cell contact, indicating a critical role of constitutive autotropic signaling of TNF.TNF receptor complexes. These data demonstrate that autotropic signaling loops of membrane TNF can exist, which may be of importance for cells that express both TNF and TNF receptors, such as T lymphocytes, macrophages, and endothelial cells.


Subject(s)
Membrane Proteins/genetics , Mitogen-Activated Protein Kinases , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspases/metabolism , Cycloheximide/pharmacology , Genes, Reporter , HeLa Cells , Humans , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Mutation , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Transfection , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases
16.
J Rheumatol ; 26(3): 645-53, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10090177

ABSTRACT

OBJECTIVE: Articular cartilage is the main target for tumor necrosis factor-alpha (TNF-alpha) and interleukin 1(IL-1) actions. These cytokines are believed to mediate cartilage degradation in arthritis. We studied the expression of TNF receptors (TNF-R) on human articular chondrocytes and their regulation by IL-1beta, TNF-alpha, and basic fibroblast growth factor (bFGF). METHODS: The expression of TNF-R55 and TNF-R75 on human nonarthritic articular chondrocytes was analyzed on protein and mRNA levels by ligand binding studies and reverse transcription polymerase chain reaction (RT-PCR) technique. The regulation of these receptors induced by IL-1 TNF-alpha, and bFGF on mRNA level was studied using RT-PCR. RESULTS: Both TNF-R55 and TNF-R75 are expressed constitutively on human articular chondrocytes, and the number of both receptors varied between 822 and 3880 receptors per cell, depending on the donor cartilage used. Using TNF receptor-specific antibodies, we show that normal chondrocytes express mainly TNF-R55. These results are consistent with the mRNA data obtained by RT-PCR. mRNA expression of TNF receptors is regulated by IL-1beta, TNF-alpha, and bFGF. On human chondrocytes the expression of TNF-R75 mRNA was markedly upregulated by IL-ID, TNF-alpha, and bFGF, whereas the expression of TNF-R55 mRNA remained largely unchanged. A combination of IL-1beta and TNF-alpha, but not of IL-1beta and bFGF, showed an additive effect on TNF-R75 mRNA expression. CONCLUSION: The expression of TNF-R55 and TNF-R75 on human articular chondrocytes is modulated independently by IL-1beta, TNF-alpha, and bFGF, suggesting a role of these regulatory mechanisms in the degradation processes of human articular cartilage in inflammatory joint diseases.


Subject(s)
Antigens, CD/biosynthesis , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Fibroblast Growth Factor 2/pharmacology , Interleukin-1/pharmacology , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Antigens, CD/genetics , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Chondrocytes/metabolism , DNA Primers/chemistry , Humans , RNA, Messenger/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Up-Regulation
17.
J Biol Chem ; 273(49): 33091-8, 1998 Dec 04.
Article in English | MEDLINE | ID: mdl-9830064

ABSTRACT

In this study we show that TRAIL (tumor necrosis factor-related apoptosis-inducing ligand), also called Apo2L, activates the c-Jun N-terminal kinase (JNK). Interestingly, TRAIL-induced JNK activation occurs in a cell type-specific manner. In HeLa cells, TRAIL-induced JNK activation can be completely blocked with the cysteine protease inhibitor zVAD-fmk, whereas the same inhibitor has no, or even a stimulatory, effect on JNK activation in Kym-1 cells. Hence, TRAIL can engage at least two independent pathways leading to JNK activation, one that is cysteine protease-dependent and one that is cysteine protease-independent. To investigate whether the cysteine protease-dependent signaling of TRAIL leading to JNK activation is related to the apoptotic pathway engaged by this ligand, we investigated HeLa cells stably overexpressing a dominant negative mutant of FADD (Fas-associating protein with death domain) (GFP(green fluorescent protein)DeltaFADD). In these cells, TRAIL-induced cell death and activation of the apoptosis executioner caspase-8 (FLICE/MACH) and caspase-3 (YAMA, CPP-32, Apopain), that belong to caspase subfamily of cysteine proteases, were abrogated, whereas JNK activation remained unaffected and was still sensitive toward z-VAD-fmk. Similar data were found in HeLa cells overexpressing Apo1/Fas and GFPDeltaFADD upon stimulation with agonistic antibodies. These data suggest that cross-linking of the TRAIL receptors and Apo1/Fas, respectively, engages a FADD-dependent pathway leading to the activation of apoptotic caspases and, in parallel, a FADD-independent pathway leading to the stimulation of one or more cysteine proteases capable to activate JNK but not sufficient for the induction of cell death.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspases/metabolism , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinases , Tumor Necrosis Factor-alpha/metabolism , Apoptosis Regulatory Proteins , Base Sequence , Caspase 3 , Cell Line , DNA Primers , Enzyme Activation , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , TNF-Related Apoptosis-Inducing Ligand
18.
J Immunol ; 161(6): 3136-42, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9743381

ABSTRACT

Costimulation of TNFR80 can strongly enhance TNFR60-induced cell death. In this study, we show that this enhancement is TNFR60 selective, as neither TNF-related apoptosis-inducing ligand/Apo2 ligand-, Apo1/Fas-, ceramide-, nor daunorubicin-mediated cell death was affected by costimulation of TNFR80. We further demonstrate that TNFR-associated factor 2 (TRAF2) is critically involved in both negative and positive regulation of TNF-induced cell death. Overexpression of TRAF2 and of a TRAF2 mutant, deficient in nuclear factor-kappaB activation, selectively desensitized and enhanced, respectively, TNFR60-induced cell death in HeLa cells. However, upon costimulation of TNFR80, which mediates activation of nuclear factor-kappaB and the c-Jun amino-terminal kinase via TRAF2, TNF-induced cell death is drastically enhanced in parental and TRAF2-transfected, but not in TRAF2 (87-501)-transfected cells. These data point to a critical role of TRAF2 in the apoptotic TNFR cross talk, whereby the TNFR80-dependent enhancement of TNFR60-induced cell death is due to TNFR80-mediated negative regulation of TRAF2 function(s). An interference with TRAF2 function was confirmed independently by analysis of c-Jun amino-terminal kinase activation via TNFR60 upon prestimulation of TNFR80. We propose that the apoptotic TNFR cross talk is based on TNFR80-mediated abrogation of antiapoptotic TRAF2-dependent signaling pathways initiated by TNFR60, but not Apo1/Fas or the apoptotic TNF-related apoptosis-inducing ligand receptors.


Subject(s)
Adjuvants, Immunologic/physiology , Antigens, CD/physiology , Carrier Proteins/physiology , HeLa Cells/immunology , Mitogen-Activated Protein Kinases , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins , Antigens, CD/chemistry , Antigens, CD/metabolism , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Death/drug effects , Cell Death/immunology , Ceramides/pharmacology , Daunorubicin/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/immunology , HeLa Cells/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , Ligands , Membrane Glycoproteins/physiology , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , TNF Receptor-Associated Factor 2 , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology
19.
J Clin Invest ; 101(10): 2278-89, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9593784

ABSTRACT

We investigated the effect of the acute promyelocytic leukemia (APL) specific PML/RARalpha fusion protein on the sensitivity to TNF-alpha-mediated apoptosis. The U937 leukemia cell line was transduced with PML/RARalpha cDNA. PML/RARalpha expression caused a markedly reduced sensitivity to TNF-alpha, even if apoptosis was triggered by agonistic antibodies to TNF-alpha receptors I and II (TNF-alphaRI, II). PML/RARalpha induced a 10-20-fold decrease of the TNF-alpha-binding capacity via downmodulation of both TNF-alphaRI and TNF-alphaRII: this may mediate at least in part the reduced sensitivity to TNF-alpha. Furthermore, the fusion protein did not modify Fas expression (CD95) or sensitivity to Fas-mediated apoptosis. The pathophysiological significance of these findings is supported by two series of observations. (a) Fresh APL blasts exhibit no TNF-alpha binding and are resistant to TNF-alpha-mediated apoptosis. Conversely, normal myeloblasts-promyelocytes show marked TNF-alphaR expression and are moderately sensitive to TNF-alpha-mediated cytotoxicity. Similarly, blasts from other types of acute myeloid leukemia (AML M1, M2, and M4 FAB types) show an elevated TNF-alpha binding. (b) The NB4 APL cell line, which is PML/RARalpha+, shows low TNF-alphaR expression capacity and is resistant to TNF-alpha-triggered apoptosis; conversely a PML/RARalpha- NB4 subclone (NB4.306) exhibits detectable TNF-alpha-binding capacity and is sensitive to TNF-alpha-mediated cytotoxicity. These studies indicate that the PML/RARalpha fusion protein protects against TNF-alpha-induced apoptosis, at least in part via downmodulation of TNF-alphaRI/II: this phenomenon may play a significant role in APL, which is characterized by prolonged survival of leukemic blasts.


Subject(s)
Apoptosis/physiology , Leukemia, Promyelocytic, Acute/metabolism , Neoplasm Proteins/genetics , Nuclear Proteins , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/physiology , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/toxicity , Antigens, CD/analysis , Antigens, CD/immunology , Cell Division/drug effects , Cell Division/genetics , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Promyelocytic Leukemia Protein , Protein Binding/drug effects , RNA, Messenger/analysis , Receptors, Tumor Necrosis Factor/agonists , Receptors, Tumor Necrosis Factor/immunology , Retinoic Acid Receptor alpha , Transfection , Tumor Cells, Cultured , Tumor Suppressor Proteins , fas Receptor/metabolism
20.
Eur J Immunol ; 28(1): 257-63, 1998 01.
Article in English | MEDLINE | ID: mdl-9485205

ABSTRACT

Tumor necrosis factor (TNF) mediates its biological effects by binding to two distinct but homologous receptor molecules. The type 1 receptor (TNF-R1) has been shown to be essential and sufficient for most cellular responses to soluble TNF. In contrast, only limited data exist concerning the role of the type 2 receptor (TNF-R2) in TNF responses, both in vitro and in vivo. Here, we demonstrate by the use of thymocytes from TNF-R-deficient mice that the TNF-R2-dependent enhancement of proliferation and secretion of granulocyte-macrophage colony-stimulating factor is in fact mediated by TNF-R2 on its own, independent of co-expression and/or stimulation of TNF-R1.


Subject(s)
Antigens, CD/physiology , Lymphocyte Activation , Receptors, Tumor Necrosis Factor/physiology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Concanavalin A/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombinant Proteins/pharmacology , T-Lymphocytes/metabolism , Thymus Gland/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...