Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Cell ; 6(2): 395-407, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10983986

ABSTRACT

Human T cell prolymphocytic leukemia can result from chromosomal translocations involving 14q32.1 or Xq28 regions. The regions encode a family of protooncogenes (TCL1, MTCP1, and TCL1b) of unknown function. In yeast two-hybrid screening, we found that TCL1 interacts with Akt. All TCL1 isoforms bind to the Akt pleckstrin homology domain. Both in vitro and in vivo TCL1 increases Akt kinase activity and as a consequence enhances substrate phosphorylation. In vivo, TCL1 stabilizes the mitochondrial transmembrane potential and enhances cell proliferation and survival. In vivo, TCL1 forms trimers, which associate with Akt. TCL1 facilitates the oligomerization and activation of Akt. Our data show that TCL1 is a novel Akt kinase coactivator, which promotes Akt-induced cell survival and proliferation.


Subject(s)
Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/metabolism , Cell Death , Cell Division , Cell Line , Chromosomes, Human, Pair 14 , Cloning, Molecular , Glycogen Synthase Kinase 3 , Humans , Intracellular Membranes/physiology , Lymphocyte Activation , Membrane Potentials , Mitochondria/physiology , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogenes , Recombinant Proteins/metabolism , Spodoptera , T-Lymphocytes/immunology , Transfection , Translocation, Genetic , Tumor Cells, Cultured , X Chromosome , bcl-Associated Death Protein , src Homology Domains
2.
J Exp Med ; 191(11): 1975-85, 2000 Jun 05.
Article in English | MEDLINE | ID: mdl-10839812

ABSTRACT

Hepatocyte apoptosis is crucial in several forms of liver disease. Here, we examined in different models of murine liver injury whether and how metabolically induced alterations of hepatocyte ATP levels control receptor-mediated apoptosis. ATP was depleted either in primary hepatocytes or in vivo by various phosphate-trapping carbohydrates such as fructose. After the activation of the tumor necrosis factor (TNF) receptor or CD95, the extent of hepatocyte apoptosis and liver damage was quantified. TNF-induced cell death was completely blocked in ATP-depleted hepatocyte cultures, whereas apoptosis mediated by CD95 was enhanced. Similarly, acute TNF-induced liver injury in mice was entirely inhibited by ATP depletion with ketohexoses, whereas CD95-mediated hepatotoxicity was enhanced. ATP depletion prevented mitochondrial cytochrome c release, loss of mitochondrial membrane potential, activation of type II caspases, DNA fragmentation, and cell lysis after exposure to TNF. The extent of apoptosis inhibition correlated with the severity of ATP depletion, and TNF-induced apoptosis was restored when ATP was repleted by increasing the extracellular phosphate concentration. Our study demonstrates that TNF-induced hepatic apoptosis can be selectively and reversibly blocked upstream of mitochondrial dysfunction by ketohexose-mediated ATP depletion.


Subject(s)
Adenosine Triphosphate/metabolism , Antigens, CD/metabolism , Apoptosis , Fructose/metabolism , Liver/cytology , Receptors, Tumor Necrosis Factor/metabolism , fas Receptor/metabolism , Animals , Caspases/metabolism , Cells, Cultured , Cycloheximide/pharmacology , Cytochrome c Group/metabolism , Enzyme Activation , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C , Protein Synthesis Inhibitors/pharmacology , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
3.
Hepatology ; 30(5): 1241-51, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10534346

ABSTRACT

Several models of tumor necrosis factor (TNF)/TNF-receptor 1 (TNF-R1)-dependent liver injury in mice were investigated with respect to caspase-3-like protease activation representing a pivotal mechanism of apoptotic cell death. Injection of TNF or T-cell-activating agents (i.e., agonistic anti-CD3 antibody or staphylococcal enterotoxin B [SEB]) into galactosamine (GalN)-sensitized mice caused TNF/TNF-R1-dependent liver injury. Intravenous concanavalin A (Con A) alone induced TNF-mediated hepatotoxicity dependent on both TNF-R1 and TNF-R2. Hepatic caspase-3-like proteases were activated in GalN/TNF, GalN/anti-CD3, or GalN/SEB-treated mice, but not in Con A-treated mice. Consistently, the broad-spectrum caspase inhibitor, benzoyloxycarbonyl-val-ala-asp-fluoromethylketone (zVADfmk), prevented TNF-mediated hepatotoxicity in all GalN-dependent models, but failed to protect against Con A. Under transcriptional arrest, however, Con A induced TNF-R1-dependent, but not TNF-R2-dependent, activation of caspase-3-like proteases, and zVADfmk prevented animals from Con A-mediated liver injury under this condition. Histological analysis revealed distinct differences between Con A- and GalN/Con A-induced liver injury regarding apoptotic morphology of hepatocytes. We conclude that impaired transcription induces a switch of Con A hepatotoxicity toward a caspase-3-like protease-dependent pathway. The observation that the functional state of the transcriptional machinery decides whether TNF-driven hepatocyte apoptosis involves activation of caspase-3-like proteases or alternative signaling pathways in vivo might be of relevance for the immunopathology of the liver.


Subject(s)
Caspases/metabolism , Concanavalin A/toxicity , Liver/pathology , Tumor Necrosis Factor-alpha/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antibodies/pharmacology , Antigens, CD/physiology , Apoptosis/drug effects , CD3 Complex/physiology , Caspase 3 , Cysteine Proteinase Inhibitors/pharmacology , Enterotoxins/toxicity , Enzyme Activation , Galactosamine/toxicity , Lipid Metabolism , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred BALB C , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Specific Pathogen-Free Organisms , Staphylococcus aureus
4.
Hepatology ; 30(1): 177-85, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385654

ABSTRACT

Agonistic engagement of the cytokine receptor CD95 in mice leads to activation of hepatic caspases, followed by massive hepatocyte apoptosis, acute liver failure, and death. This mechanism of cell death is thought to be associated with several human liver disorders. Because hepatic glutathione represents the major defense against toxic liver injury, we investigated its role in CD95-mediated liver failure, which represents a model for hyperinflammatory organ destruction. As a tool for modulating the liver glutathione status of mice in vivo, we used the GSH transferase substrate, phorone, which rapidly depleted hepatic glutathione in a dose-dependent manner. When GSH was depleted, CD95-initiated hepatic caspase-3-like activity and DNA fragmentation were completely blocked, and animals were protected from liver injury dose-dependently as assessed by histological examination and determination of liver enzymes in plasma. Conversely, repletion of hepatic glutathione by treatment with the permeable glutathione monoethylester restored susceptibility of GSH-depleted mice toward CD95-mediated liver injury. In contrast, the antioxidants, GSH, N-acetyl cysteine, alpha-tocopherol, butyl-hydroxytoluene, and catalase failed to do so. Animals treated once with phorone survived for more than 3 months after an otherwise lethal injection of the activating anti-CD95 antibody. We investigated the thiol sensitivity of recombinant caspase-3 in vitro and observed that its activity was dependent on the presence of a reducing agent such as GSH, while GSSG attenuated proteolytic activity. Based on our finding that CD95-mediated hepatocyte apoptosis requires an intact intracellular glutathione status, we propose that the activation of apoptosis-executing caspases is controlled by reduced glutathione.


Subject(s)
Apoptosis/physiology , Glutathione/metabolism , Ketones/pharmacology , Liver/cytology , fas Receptor/physiology , Acetylcysteine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Butylated Hydroxytoluene/pharmacology , Caspase 3 , Caspases/metabolism , Catalase/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , Glutathione/antagonists & inhibitors , Glutathione Disulfide/metabolism , Glutathione Transferase/metabolism , Humans , Liver/pathology , Liver/physiology , Male , Mice , Mice, Inbred BALB C , Vitamin E/pharmacology , fas Receptor/immunology
5.
J Pharmacol Exp Ther ; 287(3): 1098-104, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9864298

ABSTRACT

The seleno-organic drug ebselen (2-phenyl-1, 2-benzoisoselenazol-3(2H)-one) has glutathione peroxidase-like activity, and inhibits lipoxygenases, oxidative burst of leukocytes, nitric oxide synthases, protein kinases and leukocyte migration. This study elaborates in vivo in mice hitherto unknown immunopharmacological properties of ebselen. The compound was comparatively investigated in two different T cell-dependent hepatic hyperinflammation models and in two alternative models of receptor-activated liver apoptosis. Mice orally pretreated with ebselen were dose-dependently protected from concanavalin A (ConA)-induced liver injury. In livers from ebselen-pretreated mice exposed to ConA, the nuclear antiapoptotic transcription factor NFkappaB was upregulated. The release of the proinflammatory cytokine tumor necrosis factor-alpha (TNF) was downregulated, while the ciculating amount of the anti-inflammatory cytokine interleukin-10 (IL-10) was increased. Ebselen protected also from liver injury induced by the superantigen staphylococcal enterotoxin B in galactosamine (GalN)-sensitized mice. Furthermore, ebselen protected the liver and enhanced circulating IL-10 in GalN-sensitized mice treated with recombinant TNF, i.e., the common distal mediator of ConA and SEB-induced hepatotoxicity. The activation of apoptosis-executing proteases, i.e., caspases, was blocked in livers of ebselen-treated mice following TNF receptor, but not following CD95 receptor activation. We propose a novel mechanism for the immunomodulatory properties of the drug and suggest that it might be useful in the therapy of T cell-mediated inflammatory disorders.


Subject(s)
Azoles/pharmacology , Immunosuppressive Agents/pharmacology , Liver/drug effects , Organoselenium Compounds/pharmacology , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Caspases/metabolism , Concanavalin A , Enterotoxins , Enzyme Activation , Isoindoles , Liver/metabolism , Liver/pathology , Liver Failure/prevention & control , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
6.
J Histochem Cytochem ; 46(10): 1175-83, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9742073

ABSTRACT

Tumor necrosis factor (TNF) induces apoptotic death of hepatocytes in the galactosamine (GalN)-sensitized mouse liver after 5 hr. In our study, the most remarkable sign of the early stage of apoptosis was the focal rupture of the outer mitochondrial membrane. Parts of the inner membrane extended through the gap of the outer membrane, whereas the rest of the inner membrane still formed the cristae. This feature appeared in hepatocytes before chromatin condensation. With the diaminobenzidine technique for localization of cytochrome oxidase activity, the reaction product was detectable by light and electron microscopy. Ten percent of the hepatocytes were apoptotic, with condensed chromatin and high enzyme activity, 37% were pre-apoptotic, without chromatin condensation but high enzyme activity, and 53% had neither condensed chromatin nor a remarkable reaction product of cytochrome oxidase activity. Fas (APO-1, CD95) molecules on the plasma membrane of hepatocytes increased and were represented immunohistochemically in cells without chromatin condensation. DNA strand breaks were also detectable before chromatin aggregation. The results of this study indicate that mitochondria play a pivotal role in pre-apoptotic hepatocytes, together with an increase of the Fas molecule on the plasma membrane and with the occurrence of DNA strand breaks in the nucleus.


Subject(s)
Apoptosis , Galactosamine/pharmacology , Liver/pathology , Liver/ultrastructure , Tumor Necrosis Factor-alpha/pharmacology , Animals , Chromatin/pathology , DNA Fragmentation , Electron Transport Complex IV/analysis , Histocytochemistry , Immunohistochemistry , Liver/chemistry , Liver/drug effects , Male , Mice , Mice, Inbred BALB C , Mitochondria/pathology , fas Receptor/analysis
8.
Eur J Immunol ; 27(11): 2870-5, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9394812

ABSTRACT

The significance of tumor necrosis factor receptor 1 (TNFR1) for TNF function in vivo is well documented, whereas the role of TNFR2 so far remains obscure. In a model of concanavalin A (Con A)-induced, CD4+ T cell-dependent experimental hepatitis in mice, in which TNF is a central mediator of apoptotic and necrotic liver damage, we now provide evidence for an essential in vivo function of TNFR2 in this pathophysiological process. We demonstrate that a cooperation of TNFR1 and TNFR2 is required for hepatotoxicity as mice deficient of either receptor were resistant against Con A. A significant role of TNFR2 for Con A-induced hepatitis is also shown by the enhanced sensitivity of transgenic mice overexpressing the human TNFR2. The ligand for cytotoxic signaling via both TNF receptors is the precursor of soluble TNF, i.e. transmembrane TNF. Indeed, transmembrane TNF is sufficient to mediate hepatic damage, as transgenic mice deficient in wild-type soluble TNF but expressing a mutated nonsecretable form of TNF developed inflammatory liver disease.


Subject(s)
Hepatitis, Animal/immunology , Membrane Proteins/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Concanavalin A/toxicity , Hepatitis, Animal/chemically induced , Hepatitis, Animal/genetics , Immunity, Innate , Interferon-gamma/biosynthesis , Interferon-gamma/drug effects , Liver Failure, Acute/chemically induced , Liver Failure, Acute/genetics , Liver Failure, Acute/immunology , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Tumor Necrosis Factor-alpha/metabolism
9.
Biochem Biophys Res Commun ; 233(2): 518-22, 1997 Apr 17.
Article in English | MEDLINE | ID: mdl-9144569

ABSTRACT

Cleavage of poly-(ADP-ribose) polymerase is a process occurring early during the execution phase of apoptosis. Although in many experimental systems PARP cleavage indicates a point of no return, the significance of this proteolytic step for apoptosis remains unclear. Here we compare the susceptibility of cells from wild-type mice and PARP-/- mice to several inducers of apoptosis. Neither the susceptibility of hepatocytes towards CD95 or TNF-mediated apoptosis nor the activation of PARP-cleaving caspases was modified in PARP-/- liver cells. Thymocytes with either genotype exhibited similar sensitivity to treatments with ceramide, dexamethasone, or etoposide. The sensitivity of primary neurons towards apoptosis induced by staurosporine, colchicine, potassium withdrawal, peroxynitrite, or the neurotoxin MPP+ was also unaltered. These data suggest that neither activation nor cleavage of PARP has a causal role in apoptotic cell death of primary, non-transformed cells.


Subject(s)
Apoptosis , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cells, Cultured , Electrophoresis, Agar Gel , L-Lactate Dehydrogenase/metabolism , Liver/metabolism , Mice , Microscopy, Fluorescence , Tumor Necrosis Factor-alpha/metabolism
10.
Immunol Lett ; 55(1): 5-10, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9093874

ABSTRACT

The two apoptosis receptors of mammalian cells, i.e. the 55 kDa TNF receptor (TNF-R1) and CD95 (Fas/APO1) are activated independently of each other, however, their signaling involves a variety of ICE-related proteases [I]. We used a cell-permeable inhibitor of ICE-like protease activity to examine in vivo whether post-receptor signaling of TNF and CD95 are fully independent processes. Mice pretreated with the inhibitor, Z-VAD-fluoromethylketone (FMK) were dose-dependently protected from liver injury caused by CD95 activation as determined by plasma alanine aminotransferase and also from hepatocyte apoptosis assessed by DNA fragmentation (ID50 = 0.1 mg/kg). A dose of 10 mg/kg protected mice also from liver injury induced by TNF-alpha. Similar results were found when apoptosis was initiated via TNF-alpha or via CD95 in primary murine hepatocytes (IC50 = 1.5 nM) or in various human cell lines. In addition to prevention, an arrest of cell death by Z-VAD-FMK was demonstrated in vivo and in vitro after stimulation of apoptosis receptors. These findings show in vitro and in vivo in mammals that CD95 and the TNF-alpha receptor share a distal proteolytic apoptosis signal.


Subject(s)
Amino Acid Chloromethyl Ketones/therapeutic use , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Cysteine Endopeptidases/physiology , Cysteine Proteinase Inhibitors/therapeutic use , Liver/drug effects , Tumor Necrosis Factor-alpha/toxicity , fas Receptor/physiology , Alanine Transaminase/blood , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antigens, CD/drug effects , Antigens, CD/physiology , Carcinoma, Hepatocellular/pathology , Caspase 1 , Cells, Cultured , Chemical and Drug Induced Liver Injury/physiopathology , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , HeLa Cells/drug effects , Humans , Interleukin-1/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Lipopolysaccharides , Liver/cytology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Tumor Cells, Cultured
11.
Gastroenterology ; 111(2): 462-71, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8690213

ABSTRACT

BACKGROUND & AIMS: T cell-dependent liver injury involving endogenous tumor necrosis factor (TNF) alpha can be induced by either concanavalin A in naive mice or by activating anti-CD3 antibody or staphylococcal enterotoxin B in D-galactosamine-sensitized mice. In this study, the role of interferon gamma (IFN-gamma) in these T-cell models was addressed. METHODS: Mice were pretreated with a neutralizing anti-mouse IFN-gamma antiserum before injection of T cell-activating agents. Plasma cytokine and transaminase levels were determined. Apoptotic cell death was assessed by hepatic DNA fragmentation. RESULTS: Anti-IFN-gamma antiserum significantly protected mice from concanavalin A-induced liver injury. Circulating IFN-gamma was completely suppressed, and TNF was reduced by 50%. Recombinant TNF-alpha administered to mice treated with concanavalin A and anti-IFN-gamma antiserum failed to initiate liver injury. Similar results were obtained with recombinant IFN-gamma in concanavalin A-challenged mice under the condition of TNF neutralization. Neither hepatic DNA fragmentation nor release of transaminases was inhibited by anti-IFN-gamma antiserum when liver injury was induced by staphylococcal enterotoxin B or anti-CD3 antibody in D-galactosamine-sensitized mice. CONCLUSIONS: Both TNF as well as IFN-gamma are critical mediators of liver injury in concanavalin A-treated mice, whereas hepatic DNA fragmentation and liver failure in the D-galactosamine models depend only on TNF.


Subject(s)
Concanavalin A/adverse effects , Interferon-gamma/physiology , Liver/pathology , T-Lymphocytes/physiology , Animals , Apoptosis , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , DNA Damage , Enterotoxins/adverse effects , Enzyme-Linked Immunosorbent Assay , Galactosamine/adverse effects , Immune Sera/pharmacology , Interferon-gamma/blood , Interferon-gamma/immunology , Liver/drug effects , Liver/metabolism , Liver Failure/chemically induced , Liver Failure/metabolism , Liver Failure/pathology , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Proteins/pharmacology , Transaminases/blood , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology
12.
Mol Med ; 2(1): 109-24, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8900539

ABSTRACT

BACKGROUND: Activation of either the 55-kD tumor necrosis factor receptor (TNF-R1) or CD95 (Fas/Apo-1) causes apoptosis of cells and liver failure in mice, and has been associated with human liver disorders. The aim of this study was first to clarify the association between CD95 activation, hepatocyte apoptosis, and fulminant liver failure. Next, we investigated whether TNF-R1 and CD95 operate independently of each other in the induction of hepatocyte apoptosis. MATERIALS AND METHODS: Using both mice and primary liver cell cultures deficient in either TNF-R1 or functional CD95, the induction of apoptosis and hepatocyte death following activation of TNF-R1 or CD95 were studied in vitro and in various in vivo models of acute liver failure. RESULTS: In vivo or in vitro stimulation of CD95 caused apoptosis of wild-type (wt) murine hepatocytes which had not been sensitized by blocking transcription. Time course studies showed that DNA fragmentation and chromatin condensation preceded, respectively, membrane lysis in vitro and necrosis in vivo. Similar results were obtained after CD95 activation in hepatocytes or livers lacking TNF-R1. Conversely, hepatocytotoxicity due to endogenous or exogenous TNF was not affected in animals or liver cell cultures lacking the expression of functional CD95. CONCLUSIONS: TNF-R1 and CD95 are independent and differentially regulated triggers of murine apoptotic liver failure.


Subject(s)
Apoptosis/genetics , Liver/metabolism , Receptors, Tumor Necrosis Factor/metabolism , fas Receptor/metabolism , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Cells, Cultured , Concanavalin A/pharmacology , DNA Damage/drug effects , DNA Damage/genetics , Dactinomycin/pharmacology , Electrophoresis, Agar Gel , Galactosamine/pharmacology , Gene Expression/genetics , L-Lactate Dehydrogenase/metabolism , Liver/cytology , Liver/injuries , Lymphotoxin-alpha/pharmacology , Mice , Mice, Inbred BALB C , Models, Biological , Oxygen/metabolism , Receptors, Tumor Necrosis Factor/deficiency , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/immunology , fas Receptor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...