Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Clin Exp Immunol ; 188(1): 63-78, 2017 04.
Article in English | MEDLINE | ID: mdl-27880975

ABSTRACT

Systemic lupus erythematosus (SLE) is a common autoimmune disorder with a complex and poorly understood immunopathogenesis. However, a pathogenic role for the T helper type 17 (Th17) axis was demonstrated by many studies, while regulatory T cells (Tregs ) were shown to mediate protection. Recently, we and others characterized a novel and independent T cell population expressing both the Treg characteristic transcription factor forkhead box protein 3 (FoxP3) and the Th17-defining retinoic acid receptor-related orphan nuclear receptor γt (RORγt). Studies in a model of acute glomerulonephritis unveiled potent regulatory, but also proinflammatory, functions of RORγt+ FoxP3+ Tregs . This bi-functional nature prompted us to suggest the name 'biTregs '. Importantly, the pathogenic biTreg effects were dependent upon expression of RORγt. We thus aimed to evaluate the contribution of RORγt+ FoxP3+ biTregs to pristane-induced SLE and explored the therapeutic potential of interference with RORγt activation. Our analyses revealed expansion of IL-17 producing biTregs in a distinctive time-course and organ-specific pattern, coincident with the development of autoimmunity and tissue injury. Importantly, specific ablation of RORγt activation in endogenous biTregs resulted in significant amelioration of pristane-induced pulmonary vasculitis and lupus nephritis. As potential mechanisms underlying the observed protection, we found that secretion of IL-17 by biTregs was abrogated completely in FoxP3Cre  × RORCfl/fl mice. Furthermore, Tregs showed a more activated phenotype after cell-specific inactivation of RORγt signalling. Finally, and remarkably, biTregs were found to potently suppress anti-inflammatory Th2 immunity in a RORγt-dependent manner. Our study thus identifies biTregs as novel players in SLE and advocates RORγt-directed interventions as promising therapeutic strategies.


Subject(s)
Gene Expression , Interleukin-17/metabolism , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Immunity, Humoral , Immunomodulation , Immunophenotyping , Lung/metabolism , Lung/pathology , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/etiology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Lymphocyte Count , Male , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
2.
Eur J Pain ; 19(7): 953-65, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25429980

ABSTRACT

Acetaminophen/paracetamol is the most widely used drug of the world. At the same time, it is probably one of the most dangerous compounds in medical use, causing hundreds of deaths in all industrialized countries due to acute liver failure (ALF). Publications of the last 130 years found in the usual databases were analyzed. Personal contacts existed to renowned researchers having contributed to the medical use of paracetamol and its precursors as H.U. Zollinger, S. Moeschlin, U. Dubach, J. Axelrod and others. Further information is found in earlier reviews by Eichengrün, Rodnan and Benedek, Sneader, Brune; comp. references. The history of the discovery of paracetamol starts with an error (active against worms), continues with a false assumption (paracetamol is safer than phenacetin), describes the first side-effect 'epidemy' (phenacetin nephropathy, drug-induced interstitial nephritis) and ends with the discovery of second-generation problems due to the unavoidable production of a highly toxic metabolite of paracetamol N-acetyl-p-benzoquinone imine (NAPQI) that may cause not only ALF and kidney damage but also impaired development of the fetus and the newborn child. It appears timely to reassess the risk/benefit ratio of this compound.


Subject(s)
Acetaminophen/adverse effects , Acetaminophen/therapeutic use , Analgesics, Non-Narcotic/adverse effects , Analgesics, Non-Narcotic/therapeutic use , Acetaminophen/history , Acetaminophen/pharmacology , Adult , Analgesics, Non-Narcotic/history , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Child , History, 19th Century , History, 20th Century , History, 21st Century , Humans
4.
Z Gastroenterol ; 50(1): 34-40, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22222796

ABSTRACT

Heme oxygenases (HO) are essential enzymes which degrade heme into carbon monoxide (CO), biliverdin and free iron. Due to its anti-inflammatory, anti-apoptotic and, as recently described, anti-viral properties the inducible HO isoform HO-1 is an important molecule which could find its way into therapy of gastrointestinal diseases. Acute and chronic liver injuries including acute liver failure, alcoholic or viral hepatitis, chronic inflammation, fibrosis, cirrhosis, and hepatocellular carcinoma are life threatening diseases and as a consequence might result in the necessity of liver transplantation. HO-1 as well as its reaction products of heme degradation has been linked to cytoprotection. HO-1 induction in rodent models of acute and chronic hepatic inflammation resulted in improvement of liver damage and down-regulation of pro-inflammatory cytokine levels. Furthermore HO-1 induction interfered with fibrosis progression in mice and partially resolved existing fibrosis. Likewise, HO-1 induction interfered with replication of hepatitis viruses B and C, which frequently are the reason for chronic hepatitis and subsequent tumor growth. Liver transplantation is limited by ischemia/reperfusion (I/R) injury, which is characterized by hypoxia and nutrient deficiency resulting in oxidative stress, apoptosis and immune activation. Induction of HO-1 and application predominantly of CO have been shown to interfere with I/R liver injury and to improve recipient and graft survival. On the other hand HO-1 has been shown to be over-expressed in various tumors, including hepatocellular carcinoma (HCC). Due to its anti-apoptotic properties this bears the risk to promote tumor growth. Anti-apoptotic effects are predominantly mediated by CO. This review aims to summarize beneficial as well as detrimental effects of HO-1 and its products within the liver.


Subject(s)
Heme Oxygenase-1/metabolism , Liver Diseases/enzymology , Liver/enzymology , Animals , Humans , Mice , Rats
5.
Gut ; 58(12): 1670-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19470497

ABSTRACT

BACKGROUND AND AIMS: The transcription factor nuclear factor kappa B (NF-kappaB) has risen as a promising target for anti-inflammatory therapeutics. In the liver, however, NF-kappaB inhibition mediates both damaging and protective effects. The outcome is deemed to depend on the liver cell type addressed. Recent gene knock-out studies focused on the role of NF-kappaB in hepatocytes, whereas the role of NF-kappaB in Kupffer cells has not yet been investigated in vivo. Here we present a novel approach, which may be suitable for clinical application, to selectively target NF-kappaB in Kupffer cells and analyse the effects in experimental models of liver injury. METHODS: NF-kappaB inhibiting decoy oligodeoxynucleotides were loaded upon gelatin nanoparticles (D-NPs) and their in vivo distribution was determined by confocal microscopy. Liver damage, NF-kappaB activity, cytokine levels and apoptotic protein expression were evaluated after lipopolysaccharide (LPS), d-galactosamine (GalN)/LPS, or concanavalin A (ConA) challenge and partial warm ischaemia and subsequent reperfusion, respectively. RESULTS: D-NPs were selectively taken up by Kupffer cells and inhibited NF-kappaB activation. Inhibition of NF-kappaB in Kupffer cells improved survival and reduced liver injury after GalN/LPS as well as after ConA challenge. While anti-apoptotic protein expression in liver tissue was not reduced, pro-apoptotic players such as cJun N-terminal kinase (JNK) were inhibited. In contrast, selective inhibition of NF-kappaB augmented reperfusion injury. CONCLUSIONS: NF-kappaB inhibiting decoy oligodeoxynucleotide-loaded gelatin nanoparticles is a novel tool to selectively inhibit NF-kappaB activation in Kupffer cells in vivo. Thus, liver injury can be reduced in experimental fulminant hepatitis, but increased at ischaemia-reperfusion.


Subject(s)
Kupffer Cells/metabolism , Liver Failure, Acute/prevention & control , NF-kappa B/antagonists & inhibitors , Oligodeoxyribonucleotides/therapeutic use , Reperfusion Injury/prevention & control , Animals , Apoptosis , Disease Models, Animal , Gelatin/metabolism , Genetic Therapy/methods , Liver/blood supply , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Male , Mice , NF-kappa B/metabolism , Nanoparticles , Oligodeoxyribonucleotides/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Translocation, Genetic
6.
Z Gastroenterol ; 45(1): 63-70, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17236122

ABSTRACT

The immune response to foreign or self antigens mediates liver damage during viral or autoimmune hepatitis. However, it now appears that also specific antigen-independent liver diseases, where liver damage has been attributed to occur from oxygen radical formation, seem to be mediated by cells of the innate and adaptive immune response. These liver disorders include alcoholic liver disease, non-alcoholic fatty liver disease or non-alcoholic steatohepatitis, and ischemia/reperfusion injury that impairs the function of liver grafts. Here it seems that breakdown of the gastrointestinal barrier might increase the concentration of bacterial toxins in the portal blood, which then activate cells of the innate immune system, e. g., Kupffer cells, but, depending on the nature of the toxin, probably also conventional T cells. Invariant NKT cells which specifically recognize glycolipid antigens were supposed to become activated during metabolic disorders related to obesity. However, both steatohepatitis as well as ischemia/reperfusion injury are associated with a Th1 cytokine response characterized by IFNgamma and TNFalpha elevation, that might reflect an NKT cell response on the one hand, but also conventional T lymphocytes, in particular CD4 (+) T cells, are critical for the pathophysiology of these disorders. In 1992 we described a model of T cell-dependent liver injury inducible by the T cell-mitogenic lectin concanavalin A. This model of immune-mediated liver injury was intensively used to study pathophysiological immune effector mechanisms as well as cytokine signaling important for hepatocellular apoptosis, inhibition of apoptosis and regeneration. Recently it became evident that the inflammatory response in this model is regulated by specific cytokine signals as well as by immune regulator cells. The immune-regulatory functions of the liver are of particular interest with respect to the scavenger function of this organ, being continuously exposed to foreign antigenic material from the gut which should be eliminated without causing chronic disease.


Subject(s)
Cytokines/immunology , Hepatocytes/immunology , Immunity, Innate/immunology , Liver Diseases/immunology , Liver Diseases/pathology , Liver/immunology , Liver/pathology , Animals , Cell Communication/immunology , Hepatocytes/pathology , Humans , Immunologic Factors/immunology , Models, Immunological , T-Lymphocytes/immunology , T-Lymphocytes/pathology
7.
Inflamm Res ; 51(5): 229-35, 2002 May.
Article in English | MEDLINE | ID: mdl-12056510

ABSTRACT

OBJECTIVE AND DESIGN: Pretreatment with tumor necrosis factor (TNF)-alpha induces tolerance towards itself in experimental liver injury. MATERIAL AND TREATMENT: To study mechanisms of TNF tolerance we used knockout mice for either TNF-receptor-2 (TNFR-2), inducible nitric oxide (NO)-synthase (iNOS) or caspase-1 (ICE) or inhibited heme oxygenase-1 (HO-1) by treatment with zinc-protoporphyrin 9. Liver damage was induced by administration of TNF to mice sensitized with D-galactosamine (GalN). Tolerance was induced by pretreatment with low doses of TNF. METHODS: Severity of liver injury was assessed by determination of plasma transaminases and apoptosis. Time courses of intra-hepatic iNOS, interleukin-1beta (IL-1beta) and HO-1 expression after TNF treatment were measured by reverse transcription polymerase chain reaction (RT-PCR). TNF-receptor-1 (TNFR-1) expression was determined by immunofluorescent staining. RESULTS: TNF-pretreatment did not affect TNFR-1 expression in the liver and resulted in time dependent up-regulation of iNOS, IL-1beta and HO-1. TNF- pretreated TNFR-2, iNOS or ICE knockout mice were as sensitive towards GalN/TNF as the wild type, while mice with impaired HO-1 activity were even more sensitive, but tolerance was inducible in all TNF-pretreated mice. CONCLUSIONS: TNF tolerance towards GalN/TNF treatment is mediated by TNFR-1. IL-1beta, iNOS and HO-1 neither mediated TNF-tolerance nor TNF cytotoxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Chemical and Drug Induced Liver Injury/pathology , Galactosamine/toxicity , Heme Oxygenase (Decyclizing)/physiology , Interleukin-1/physiology , Liver/pathology , Nitric Oxide Synthase/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antigens, CD/drug effects , Antineoplastic Agents/toxicity , DNA Fragmentation/drug effects , Drug Tolerance , Heme Oxygenase-1 , Immunohistochemistry , Liver/drug effects , Male , Membrane Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Nitric Oxide Synthase Type II , RNA, Messenger/biosynthesis , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/toxicity
8.
J Immunol ; 167(7): 3944-52, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564813

ABSTRACT

Endothelial activation is an important feature of many inflammatory diseases and has been implicated as the cause of vascular complications in disorders such as diabetes, atherosclerosis, and transplant rejection. One of the most potent activators of the endothelium is TNF, which can also be expressed by endothelial cells, causing a permanent, autocrine stimulatory signal. To establish a model of continuous endothelial activation and to elucidate the role of endothelial derived TNF in vivo, we generated transgenic mice expressing a noncleavable transmembrane form of TNF under the control of the endothelial-specific tie2 promoter. Adult tie2-transmembrane TNF-transgenic mice developed chronic inflammatory pathology in kidney and liver, characterized by perivascular infiltration of mononuclear cells into these organs. Along with the infiltrate, an up-regulation of the adhesion molecules ICAM-1 and VCAM-1, but not E-selectin, in the endothelium was observed. Despite predisposition to chronic inflammation these mice were protected from immune-mediated liver injury in a model of Con A-induced acute hepatitis. Although the blood levels of soluble TNF and IFN-gamma were increased in transgenic animals after challenge with Con A, no damage of hepatocytes could be detected, as assessed by the lack of increase in plasma transaminase activities and the absence of TUNEL staining in the liver. We conclude that expression of transmembrane TNF in the endothelium causes continuous endothelial activation, leading to both proinflammatory and protective events.


Subject(s)
Chemical and Drug Induced Liver Injury/therapy , Endothelium, Vascular/metabolism , Inflammation/chemically induced , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology , Acute Disease , Animals , Cell Adhesion Molecules/biosynthesis , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chronic Disease , Concanavalin A , Inflammation/metabolism , Inflammation/pathology , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Transgenic , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-2 , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
9.
Hepatology ; 34(3): 535-47, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11526540

ABSTRACT

Liver resident NK1.1+ T cells are supposed to play a pivotal role in the onset of inflammatory liver injury in experimental mouse models such as concanavalin A (Con A)-induced hepatitis. These cells, expressing the adhesion receptor, CD44, are largely depleted from the liver by a single intravenous injection of low-molecular-weight fragments of hyaluronic acid (LMW-HA). Here, we report that LMW-HA pretreatment protected mice from liver injury in several models of T-cell- and macrophage-dependent, tumor necrosis factor alpha (TNF-alpha)-mediated inflammatory liver injury, i.e., from liver injury induced by either Con A or Pseudomonas exotoxin A (PEA) or PEA/lipopolysaccharide (LPS). Interestingly, apart from inhibition of cellular adhesion, pretreatment of mice with LMW-HA was also capable of preventing hepatocellular apoptosis and activation of caspase-3 induced by direct administration of recombinant murine (rmu) TNF-alpha to D-galactosamine (GalN)-sensitized mice. LMW-HA-induced hepatoprotection could be neutralized by pretreatment with the nuclear factor-kappaB (NF-kappaB) inhibitor, pyrrolidine dithiocarbamate (PDTC), demonstrating the involvement of NF-kappaB in the observed protective mechanism. Indeed, injection of LMW-HA rapidly induced the production of TNF-alpha by Kupffer cells and the translocation of NF-kappaB into hepatocellular nuclei. Both LMW-HA-induced TNF-alpha production and NF-kappaB translocation were blocked by pretreatment with PDTC. Our findings provide evidence for an unknown mechanism of LMW-HA-dependent protection from inflammatory liver disease, i.e., induction of TNF-alpha- and NF-kappaB-dependent cytoprotective proteins within the target parenchymal liver cells.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Chemical and Drug Induced Liver Injury/prevention & control , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , NF-kappa B/physiology , Tumor Necrosis Factor-alpha , Virulence Factors , Animals , CD4 Lymphocyte Count , Cell Death/drug effects , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/pharmacology , Cytokines/blood , Exotoxins/pharmacology , Hyaluronan Receptors/analysis , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Liver/pathology , Liver Failure/etiology , Liver Failure/prevention & control , Lymphocyte Count , Macrophages/physiology , Male , Mice , Mice, Inbred BALB C , Molecular Weight , NF-kappa B/antagonists & inhibitors , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/physiology , Pseudomonas aeruginosa Exotoxin A
10.
J Immunol ; 167(1): 514-23, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418690

ABSTRACT

Con A administration results in dose-dependent immune-mediated liver injury. Cytokines are important to determine the outcome of liver failure in this model, and especially TNF-alpha and IFN-gamma directly contribute to hepatocyte damage. The intracellular pathways of these two cytokines, which eventually result in tissue destruction, are not well defined. Here we used anti-IFN-gamma Abs and adenoviral vectors that express molecules inhibiting distinct TNF-alpha-dependent pathways in hepatocytes to better understand the relevance of specific intracellular signaling cascades for Con A-induced liver failure. We show that activation of TNF-alpha- and IFN-gamma-dependent intracellular pathways occurs prior to the influx of immune-activated cells into the liver and that anti-TNF-alpha and anti-IFN-gamma neutralizing Abs cannot block infiltration of these cells. Blocking experiments with Abs and adenoviral vectors showed that NF-kappaB activation and the Fas-associated death domain protein/caspase 8 cascade in hepatocytes during Con A-induced liver failure have no impact on tissue injury. Additionally, STAT1 activation alone after Con A injection in liver cells does not result in liver damage. In contrast, IFN-gamma-dependent expression of IFN regulatory factor-1 and TNF-alpha-dependent activation of c-Jun N-terminal kinase in liver cells correlates with liver cell damage after Con A injection. Therefore, our experiments indicate that 11418690


Subject(s)
Adaptor Proteins, Signal Transducing , Concanavalin A/administration & dosage , DNA-Binding Proteins/physiology , Hepatocytes/enzymology , Interferon-gamma/physiology , Intracellular Fluid/enzymology , Liver Failure/enzymology , Liver Failure/immunology , Mitogen-Activated Protein Kinases/physiology , Phosphoproteins/physiology , Animals , CD4-Positive T-Lymphocytes/pathology , Carrier Proteins/metabolism , Cell Movement/immunology , Concanavalin A/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Fas-Associated Death Domain Protein , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Immune Sera/administration & dosage , Injections, Intravenous , Interferon Regulatory Factor-1 , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Intracellular Fluid/immunology , JNK Mitogen-Activated Protein Kinases , Leukocyte Common Antigens/biosynthesis , Liver Failure/pathology , Liver Failure/prevention & control , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , STAT1 Transcription Factor , Signal Transduction/immunology , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/metabolism
11.
J Clin Invest ; 107(4): 439-47, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11181643

ABSTRACT

Concanavalin A (Con A) causes severe TNF-alpha-mediated and IFN-gamma-mediated liver injury in mice. In addition to their other functions, TNF-alpha and IFN-gamma both induce the inducible nitric oxide (NO) synthase (iNOS). Using different models of liver injury, NO was found to either mediate or prevent liver damage. To further elucidate the relevance of NO for liver damage we investigated the role of iNOS-derived NO in the Con A model. We report that iNOS mRNA was induced in livers of Con A-treated mice within 2 hours, with iNOS protein becoming detectable in hepatocytes as well as in Kupffer cells within 4 hours. iNOS-/- mice were protected from liver damage after Con A treatment, as well as in another TNF-alpha-mediated model that is inducible by LPS in D-galactosamine-sensitized (GalN-sensitized) mice. iNOS-deficient mice were not protected after direct administration of recombinant TNF-alpha to GalN-treated mice. Accordingly, pretreatment of wild-type mice with a potent and specific inhibitor of iNOS significantly reduced transaminase release after Con A or GalN/LPS, but not after GalN/TNF-alpha treatment. Furthermore, the amount of plasma TNF-alpha and of intrahepatic TNF-alpha mRNA and protein was significantly reduced in iNOS-/- mice. Our results demonstrate that iNOS-derived NO regulates proinflammatory genes in vivo, thereby contributing to inflammatory liver injury in mice by stimulation of TNF-alpha production.


Subject(s)
Liver/pathology , Nitric Oxide Synthase/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Concanavalin A/toxicity , Interferon-gamma/biosynthesis , Liver/enzymology , Lysine/analogs & derivatives , Lysine/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide Synthase Type II
12.
J Immunol ; 166(2): 1300-7, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11145713

ABSTRACT

TNF-alpha has been clearly identified as central mediator of T cell activation-induced acute hepatic injury in mice, e.g., Con A hepatitis. In this model, liver injury depends on both TNFRs, i.e., the 55-kDa TNFR1 as well as the 75-kDa TNFR2. We show in this report that the hepatic TNFRs are not transcriptionally regulated, but are regulated by receptor shedding. TNF directly mediates hepatocellular death by activation of TNFR1 but also induces the expression of inflammatory proteins, such as cytokines and adhesion molecules. Here we provide evidence that resistance of TNFR1(-/-) and TNFR2(-/-) mice against Con A hepatitis is not due to an impaired production of the central mediators TNF and IFN-gamma. Con A injection results in a massive induction of ICAM-1, VCAM-1, and E-selectin in the liver. Lack of either one of both TNFRs did not change adhesion molecule expression in the livers of Con A-treated mice, presumably reflecting the fact that other endothelial cell-activating cytokines up-regulated adhesion molecule expression. However, treatment of TNFR1(-/-) and TNFR2(-/-) mice with murine rTNF revealed a predominant role for TNFR1 for the induction of hepatic adhesion molecule expression. Pretreatment with blocking Abs against E- and P-selectin or of ICAM(-/-) mice with anti-VCAM-1 Abs failed to prevent Con A hepatitis, although accumulation of the critical cell population, i.e., CD4(+) T cells was significantly inhibited. Hence, up-regulation of adhesion molecules during acute hepatitis unlikely contributes to organ injury but rather represents a defense mechanism.


Subject(s)
Antigens, CD/physiology , Cell Adhesion Molecules/biosynthesis , Chemical and Drug Induced Liver Injury/immunology , Concanavalin A/toxicity , Liver/immunology , Liver/metabolism , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD/genetics , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/physiology , Cell Movement/immunology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/administration & dosage , Cytokines/biosynthesis , E-Selectin/biosynthesis , E-Selectin/immunology , Injections, Intraperitoneal , Injections, Intravenous , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/physiology , Liver/pathology , Liver Failure, Acute/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA, Messenger/biosynthesis , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombinant Proteins/administration & dosage , Solubility , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/physiology
13.
Am J Pathol ; 157(5): 1671-83, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073826

ABSTRACT

T cells seem to be responsible for liver damage in any type of acute hepatitis. Nevertheless, the importance of Kupffer cells (KCs) for T-cell-dependent liver failure is unclear. Here we focus on the role of KCs and tumor necrosis factor (TNF) production after T cell stimulation in mice. T-cell- and TNF-dependent liver injury were induced either by Pseudomonas exotoxin A (PEA), by concanavalin A (Con A), or by the combination of subtoxic doses of PEA and the superantigen Staphylococcus enterotoxin B (SEB). KCs were depleted by clodronate liposomes. Although livers of PEA-treated mice contained foci of confluent necrosis and numerous apoptotic cells, hardly any apoptotic cells were observed in the livers of Con A-treated mice. Instead, large bridging necroses were visible. Elimination of KCs protected mice from PEA-, Con A-, or PEA/SEB-induced liver injury. In the absence of KCs, liver damage was restricted to a few small necrotic areas. KCs were the main source of TNF. Hepatic TNF mRNA and protein production were strongly attenuated because of KC-depletion whereas plasma TNF levels were unaltered. Our results suggest that KCs play an important role in T cell activation-induced liver injury by contributing TNF. Plasma TNF levels are poor diagnostic markers for the severity of TNF-dependent liver inflammation.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Kupffer Cells/physiology , Liver Diseases/etiology , T-Lymphocytes/physiology , Virulence Factors , Animals , Antigens, Bacterial , Chemical and Drug Induced Liver Injury , Concanavalin A , Drug Combinations , Enterotoxins , Exotoxins , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Mice , Mice, Inbred BALB C , Necrosis , Tumor Necrosis Factor-alpha/biosynthesis , Pseudomonas aeruginosa Exotoxin A
14.
Immunol Lett ; 74(2): 165-72, 2000 Oct 03.
Article in English | MEDLINE | ID: mdl-10996392

ABSTRACT

Pseudomonas aeruginosa is a potentially dangerous Gram-negative nosocomial pathogen, causing bacteremia in debilitated patients, and a prominent cause of bacterial cholangitis. Opportunistic infections with other nosocomial pathogens, e.g. Staphylococcus aureus, are common. Hence, multi-intoxication with P. aeruginosa exotoxin A (PEA) and other bacterial toxins, including endotoxin (LPS) and the superantigen S. aureus enterotoxin B (SEB), is very likely. Here we show that PEA synergistically interacted with LPS, SEB, or recombinant murine tumor necrosis factor alpha (rmuTNF) in mice, resulting in severe liver injury. Enhanced and prolonged circulation of cytokines, including TNF, which depended on the presence of T cells, was a remarkable feature of synergistic PEA/LPS- or PEA/SEB-induced hepatotoxicity. PEA/LPS-, PEA/SEB- or PEA/rmuTNF-induced liver injury was mediated by both TNF receptors (TNFRs), i.e. TNFR1 and TNFR2. In view of the fact that TNFR1, but not TNFR2, signaling is unequivocally required for host defense, our results suggest that anti-TNFR2 strategies might be beneficial to protect the liver from inflammatory damage caused by synergistic interactions of PEA with other TNF-inducing bacterial toxins.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Chemical and Drug Induced Liver Injury/metabolism , Exotoxins/pharmacology , Lipopolysaccharides/pharmacology , Receptors, Tumor Necrosis Factor/physiology , Superantigens/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Virulence Factors , Alanine Transaminase/blood , Animals , Antibodies, Monoclonal/pharmacology , Cytokines/blood , DNA Fragmentation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Tumor Necrosis Factor/genetics , Staphylococcus aureus/immunology , T-Lymphocytes/drug effects , Thy-1 Antigens/immunology , Pseudomonas aeruginosa Exotoxin A
15.
Toxicology ; 138(2): 103-26, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10576587

ABSTRACT

Intoxication with different natural toxins or man-made toxicants has been associated with the induction of tumor necrosis factor alpha (TNF). These include endotoxin, superantigens, Pseudomonas aeruginosa exotoxin A, bacterial DNA, T cell stimulatory agents such as agonistic anti-CD3 mAbs or concanavalin A, alpha-amanitin, paracetamol, ethanol, carbon tetrachloride, dioxin, and dimethylnitrosamine. In this paper we compile and discuss the current knowledge on the pathophysiological role of TNF during intoxication with all mentioned toxins and toxicants. A possible role of gut-derived endotoxin in several TNF-dependent toxic events has been considered. The development of pharmaceuticals that selectively interfere with the detrimental pathways induced by TNF during intoxication with bacteria, viruses, drugs, or other chemicals requires detailed knowledge of the signaling pathways originating from the two TNF receptors (TNFR1 and TNFR2). Major characteristics of these signaling pathways are described and put together.


Subject(s)
Toxins, Biological/toxicity , Tumor Necrosis Factor-alpha/physiology , Xenobiotics/toxicity , Animals , Humans , Poisoning/metabolism , Poisoning/pathology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/biosynthesis
16.
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
17.
J Neuroimmunol ; 96(2): 131-43, 1999 May 03.
Article in English | MEDLINE | ID: mdl-10337911

ABSTRACT

To investigate the interaction between the peripheral nervous and the immune system in vivo, we used two mouse models of T cell and TNF-alpha dependent liver injury inducible by either concanavalin A or a combination of D-galactosamine and staphylococcal enterotoxin B. Mice depleted of peptidergic sensory nerve fibres by capsaicin were protected from liver injury. Moreover, TNF-alpha production was significantly reduced. Examination of the effect of catecholamines on liver injury showed that the beta2-adrenergic agonist salbutamol prevented, whereas chemical sympathectomy by 6-hydroxydopamine, deteriorated the disease. Hence, strategies reducing the activity of peptidergic sensory nerve fibres or stimulating beta2-adrenoreceptors, may be of benefit in immune-mediated liver disease.


Subject(s)
Hepatitis, Animal/physiopathology , Immune System Diseases/physiopathology , Neurons, Afferent/physiology , Receptors, Adrenergic, beta/physiology , Adrenergic beta-Agonists/pharmacology , Albuterol/pharmacology , Animals , Capsaicin/pharmacology , Catecholamines/physiology , Hepatitis, Animal/pathology , Liver/innervation , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Nerve Fibers/metabolism , Neurons, Afferent/drug effects , Sympathectomy, Chemical
18.
Ann N Y Acad Sci ; 887: 12-7, 1999.
Article in English | MEDLINE | ID: mdl-10668460

ABSTRACT

The electron microscopical studies presented here show that characteristic morphological alterations in mitochondria are a very early hallmark of the hepatocellular apoptotic program. Before chromatin condensation occurs, the outer mitochondrial membrane is focally disrupted and the inner membrane protrudes through this gap forming a hernia. The demonstration of cytochrome oxidase in mitochondria revealed a very strong activity in pre-apoptotic and apoptotic cells as well as in apoptotic bodies.


Subject(s)
Apoptosis/drug effects , Galactosamine/toxicity , Liver/drug effects , Mitochondria, Liver/ultrastructure , Tumor Necrosis Factor-alpha/pharmacology , Animals , Liver/pathology , Liver/ultrastructure , Mice , Mitochondria, Liver/drug effects
19.
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
20.
J Immunol ; 161(10): 5745-54, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9820556

ABSTRACT

The most potent virulence factor of Pseudomonas aeruginosa, its exotoxin A (PEA), inhibits protein synthesis, especially in the liver, and is a weak T cell mitogen. This study was performed to correlate hepatotoxic and possible immunostimulatory features of PEA in vivo. Injection of PEA to mice caused hepatocyte apoptosis, an increase in plasma transaminase activities, and the release of TNF, IFN-gamma, IL-2, and IL-6 into the circulation. Most strikingly, liver damage depended on T cells. Athymic nude mice or mice depleted of T cells by anti-Thy1.2 mAb pretreatment failed to develop acute hepatic failure, and survival was significantly prolonged following T cell depletion. Neutralization of TNF or lack of TNF receptors prevented liver injury. In the liver, TNF was produced by Kupffer cells before hepatocellular death occurred. After T cell depletion, Kupffer cells failed to produce TNF. Transaminase release was significantly reduced in perforin knockout mice, and it was even elevated in lpr/lpr mice. These results demonstrate that PEA induces liver damage not only by protein synthesis inhibition but also by TNF- and perforin-dependent, Fas-independent, apoptotic signals.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins/toxicity , Exotoxins/toxicity , Liver Diseases/pathology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Virulence Factors , Acute Disease , Animals , Cytokines/metabolism , Injections, Intravenous , Liver/pathology , Liver/ultrastructure , Liver Diseases/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Inbred Strains , Mice, Knockout , Mice, Nude , Pseudomonas Infections/immunology , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL
...