Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 9(6): 1584-1595, 2016 11.
Article in English | MEDLINE | ID: mdl-26883728

ABSTRACT

Although the development of a fully protective HIV vaccine is the ultimate goal of HIV research, to date only one HIV vaccine trial, the RV144, has successfully induced a weakly protective response. The 31% protection from infection achieved in the RV144 trial was linked to the induction of nonneutralizing antibodies, able to mediate antibody-dependent cell-mediated cytotoxicity (ADCC), suggestive of an important role of Fc-mediated functions in protection. Similarly, Fc-mediated antiviral activity was recently shown to play a critical role in actively suppressing the viral reservoir, but the Fc effector mechanisms within tissues that provide protection from or after infection are largely unknown. Here we aimed to define the landscape of effector cells and Fc receptors present within vulnerable tissues. We found negligible Fc receptor-expressing natural killer cells in the female reproductive and gastrointestinal mucosa. Conversely, Fc receptor-expressing macrophages were highly enriched in most tissues, but neutrophils mediated superior antibody-mediated phagocytosis. Modifications in Fc domain of VRC01 antibody increased phagocytic responses in both phagocytes. These data suggest that non-ADCC-mediated mechanisms, such as phagocytosis and neutrophil activation, are more likely to play a role in preventative vaccine or reservoir-eliminating therapeutic approaches.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/immunology , Phagocytosis/immunology , Receptors, Fc/metabolism , Adult , Antibodies, Monoclonal/immunology , Biomarkers , Broadly Neutralizing Antibodies , Cytokines/metabolism , Female , Gene Expression , HIV Antibodies/immunology , HIV Infections/prevention & control , HIV Infections/virology , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Middle Aged , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/virology , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Fc/genetics , Young Adult
2.
Cell Death Dis ; 5: e1102, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24603330

ABSTRACT

Systemic inflammatory response syndromes (SIRS) may be caused by both infectious and sterile insults, such as trauma, ischemia-reperfusion or burns. They are characterized by early excessive inflammatory cytokine production and the endogenous release of several toxic and damaging molecules. These are necessary to fight and resolve the cause of SIRS, but often end up progressively damaging cells and tissues, leading to life-threatening multiple organ dysfunction syndrome (MODS). As inflammasome-dependent cytokines such as interleukin-1ß are critically involved in the development of MODS and death in SIRS, and ATP is an essential activator of inflammasomes in vitro, we decided to analyze the ability of ATP removal to prevent excessive tissue damage and mortality in a murine LPS-induced inflammation model. Our results indeed indicate an important pro-inflammatory role for extracellular ATP. However, the effect of ATP is not restricted to inflammasome activation at all. Removing extracellular ATP with systemic apyrase treatment not only prevented IL-1ß accumulation but also the production of inflammasome-independent cytokines such as TNF and IL-10. In addition, ATP removal also prevented systemic evidence of cellular disintegration, mitochondrial damage, apoptosis, intestinal barrier disruption and even mortality. Although blocking ATP receptors with the broad-spectrum P2 purinergic receptor antagonist suramin imitated certain beneficial effects of apyrase treatment, it could not prevent morbidity or mortality at all. We conclude that removal of systemic extracellular ATP could be a valuable strategy to dampen systemic inflammatory damage and toxicity in SIRS.


Subject(s)
Adenosine Triphosphate/metabolism , Inflammation Mediators/metabolism , Systemic Inflammatory Response Syndrome/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis , Apyrase/pharmacology , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Endotoxins , Female , Hydrolysis , Inflammasomes/immunology , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/pathology , Purinergic P2 Receptor Antagonists/pharmacology , Signal Transduction , Suramin/pharmacology , Systemic Inflammatory Response Syndrome/chemically induced , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/pathology , Systemic Inflammatory Response Syndrome/prevention & control , Time Factors
3.
Neurogastroenterol Motil ; 25(5): e339-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23551931

ABSTRACT

BACKGROUND: Soluble guanylate cyclase (sGC) is the principal target of nitric oxide (NO) to control gastrointestinal motility. The consequence on nitrergic signaling and gut motility of inducing a heme-free status of sGC, as induced by oxidative stress, was investigated. METHODS: sGCß1 (H105F) knock-in (apo-sGC) mice, which express heme-free sGC that has basal activity, but cannot be stimulated by NO, were generated. KEY RESULTS: Diethylenetriamine NONOate did not increase sGC activity in gastrointestinal tissue of apo-sGC mice. Exogenous NO did not induce relaxation in fundic, jejunal and colonic strips, and pyloric rings of apo-sGC mice. The stomach was enlarged in apo-sGC mice with hypertrophy of the muscularis externa of the fundus and pylorus. In addition, gastric emptying and intestinal transit were delayed and whole-gut transit time was increased in the apo-sGC mice, while distal colonic transit time was maintained. The nitrergic relaxant responses to electrical field stimulation at 1-4 Hz were abolished in fundic and jejunal strips from apo-sGC mice, but in pyloric rings and colonic strips, only the response at 1 Hz was abolished, indicating the contribution of other transmitters than NO. CONCLUSIONS & INFERENCES: The results indicate that the gastrointestinal consequences of switching from a native sGC to a heme-free sGC, which cannot be stimulated by NO, are most pronounced at the level of the stomach establishing a pivotal role of the activation of sGC by NO in normal gastric functioning. In addition, delayed intestinal transit was observed, indicating that nitrergic activation of sGC also plays a role in the lower gastrointestinal tract.


Subject(s)
Gastrointestinal Motility/physiology , Gastroparesis/metabolism , Guanylate Cyclase/metabolism , Heme/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Disease Models, Animal , Gastric Mucosa/metabolism , Gene Knock-In Techniques , Guanylate Cyclase/chemistry , Mice , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Oxidative Stress/physiology , Receptors, Cytoplasmic and Nuclear/chemistry , Soluble Guanylyl Cyclase
4.
Int J Impot Res ; 20(3): 278-84, 2008.
Article in English | MEDLINE | ID: mdl-18059500

ABSTRACT

Soluble guanylyl cyclase (sGC) is the major effector molecule for nitric oxide (NO) and as such an interesting therapeutic target for the treatment of erectile dysfunction. To assess the functional importance of the sGCalpha(1)beta(1) isoform in corpus cavernosum (CC) relaxation, CC from male sGCalpha(1)(-/-) and wild-type mice were mounted in organ baths for isometric tension recording. The relaxation to endogenous NO (from acetylcholine, bradykinin and electrical field stimulation) was nearly abolished in the sGCalpha(1)(-/-) CC. In the sGCalpha(1)(-/-) mice, the relaxing influence of exogenous NO (from sodium nitroprusside and NO gas), BAY 41-2272 (NO-independent sGC stimulator) and T-1032 (phosphodiesterase type 5 inhibitor) were also significantly decreased. The remaining exogenous NO-induced relaxation seen in the sGCalpha(1)(-/-) mice was significantly decreased by the sGC-inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one. The specificity of the impairment of the sGC-related responses was demonstrated by the unaltered relaxations seen with forskolin (adenylyl cyclase activator) and 8-pCPT-cGMP (cGMP analog). In conclusion, the sGCalpha(1)beta(1) isoform is involved in corporal smooth muscle relaxation in response to NO and NO-independent sGC stimulators. The fact that there is still some effect of exogenous NO in the sGCalpha(1)(-/- mice suggests the contribution of (an) additional pathway(s).


Subject(s)
Guanylate Cyclase/physiology , Muscle Relaxation/physiology , Muscle, Smooth, Vascular/enzymology , Penile Erection/physiology , Penis/enzymology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , In Vitro Techniques , Isoenzymes , Male , Mice , Mice, Knockout , Nitric Oxide/physiology , Soluble Guanylyl Cyclase
5.
Curr Med Chem ; 13(4): 465-75, 2006.
Article in English | MEDLINE | ID: mdl-16475934

ABSTRACT

Leptin was originally identified as an adipocyte-derived cytokine with a key role in the regulation of the energy balance. Subsequent research revealed that leptin's biological action is not restricted to its effects on appetite and food intake, but instead has a much more pleiotropic character. There is now ample evidence that leptin has important functions in reproduction, hematopoiesis, HPA-axis endocrinology and angiogenesis. In this review we have focused on the effects of leptin in the antigen-specific immunity and in the inflammatory effector system.


Subject(s)
Immunity/physiology , Leptin/physiology , Systemic Inflammatory Response Syndrome/physiopathology , Adaptation, Physiological , Animals , Anorexia/physiopathology , Humans , Inflammation/physiopathology , Macrophages/drug effects , Macrophages/physiology , Metallothionein/biosynthesis , Monocytes/drug effects , Monocytes/physiology , Receptors, Cell Surface/physiology , Receptors, Leptin , Starvation/physiopathology , T-Lymphocytes/physiology , alpha-MSH/physiology
6.
Cell Mol Life Sci ; 62(14): 1632-40, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15990956

ABSTRACT

Tumor necrosis factor (TNF) is involved in pathologies like septic shock, inflammatory bowel disease and rheumatoid arthritis. TNF and lipopolysaccharide can incite lethal shock, in which cardiovascular collapse is centrally orchestrated by the vasodilating free radical nitric oxide (NO). However, NO synthase (NOS) inhibition causes increased morbidity and/or mortality, suggesting a dual role for NO. To investigate the potential protective role of NO during TNF shock, we treated mice with TNF with or without NOS inhibition. Experiments in endothelial- NOS- and inducible NOS-deficient mice identified inducible NOS as the source of protective NO. Distinctive TNF-induced lipid peroxidation, especially in liver and kidney, was aggravated by NOS inhibition. In addition, various antioxidant treatments and a phospholipase A2 (PLA2) inhibitor prevented sensitization by NOS inhibition. Together, these in vivo results indicate that induced NO not only causes hemodynamic collapse, but is also essential for curbing TNF-induced oxidative stress, which appears to hinge on PLA2-dependent mechanisms.


Subject(s)
Nitric Oxide/metabolism , Oxidative Stress , Shock/metabolism , Tumor Necrosis Factors/pharmacology , Aldehydes/analysis , Animals , Antioxidants/pharmacology , Butylated Hydroxyanisole/pharmacology , Catalase/pharmacology , Cyclic N-Oxides/pharmacology , Enzyme Inhibitors/pharmacology , Female , Immunohistochemistry , Injections, Intravenous , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Reactive Oxygen Species/metabolism , Shock/etiology , Shock/mortality , Spin Labels , Superoxide Dismutase/pharmacology , Survival Rate , Tumor Necrosis Factors/toxicity
7.
Gut ; 51(3): 398-404, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12171963

ABSTRACT

BACKGROUND: alpha(1)-Acid glycoprotein (alpha(1)-AGP) is an acute phase protein in most mammalian species whose concentration rises 2-5-fold during an acute phase reaction. Its serum concentration has often been used as a marker of disease, including inflammatory bowel disease (IBD). High alpha(1)-AGP levels were found to have a prognostic value for an increased risk of relapse in IBD. AIMS: To investigate a possible role for increased serum levels of alpha(1)-AGP in the development of IBD. METHODS: Dextran sodium sulphate (DSS) 2% was added to the drinking water of transgenic mice, overexpressing the rat alpha(1)-AGP gene, to induce acute colitis, thus mimicking the conditions of relapse. Clinical parameters, inflammatory parameters, and histological analyses on colon sections were performed. RESULTS: Homozygous alpha(1)-AGP-transgenic mice started losing weight and showed rectal bleeding significantly earlier than heterozygous transgenic or wild-type mice. Survival time of homozygous transgenic mice was significantly shorter compared with heterozygous and wild-type mice. The higher susceptibility of homozygous alpha(1)-AGP-transgenic mice to DSS induced acute colitis was also reflected in higher local myeloperoxidase levels, higher inflammation scores of the colon, and higher systemic levels of interleukin 6 and serum amyloid P component. Local inflammatory parameters were also significantly different in heterozygous transgenic mice compared with wild-type mice, indicating a local dosage effect. In homozygous transgenic mice, significantly higher amounts of bacteria were found in organs but IgA levels were only slightly lower than those of control mice. CONCLUSION: Sufficiently high serum levels of alpha(1)-AGP result in a more aggressive development of acute colitis.


Subject(s)
Colitis/blood , Orosomucoid/metabolism , Acute Disease , Animals , Colitis/chemically induced , Colony Count, Microbial , Dextran Sulfate , Disease Models, Animal , Female , Gastrointestinal Hemorrhage/blood , Immunoglobulin A/blood , Interleukin-6/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peroxidase/blood , Serum Amyloid P-Component/analysis , Weight Loss/drug effects
8.
Curr Drug Targets Inflamm Allergy ; 1(3): 277-89, 2002 Sep.
Article in English | MEDLINE | ID: mdl-14561193

ABSTRACT

Leptin was originally identified as an adipocyte-derived cytokine with a key role in the regulation of the energy balance. Subsequent research has, however, revealed that leptin's biological action is not restricted to its effects on appetite and food intake, but rather has a much more pleiotropic character. Evidence is now accumulating that it has important functions in reproduction, hematopoiesis, HPA-axis endocrinology and angiogenesis. In this review, we have focused on the effects of leptin in the immune system, which can be found in both the antigen-specific immunity and in the inflammatory effector system.


Subject(s)
Immunity/physiology , Leptin/physiology , Systemic Inflammatory Response Syndrome/physiopathology , Adaptation, Physiological , Animals , Humans , Immunity, Cellular/physiology , Inflammation/physiopathology , Metallothionein/biosynthesis , Neuropeptide Y/biosynthesis , Starvation/physiopathology , T-Lymphocytes/immunology
9.
J Exp Med ; 194(11): 1617-24, 2001 Dec 03.
Article in English | MEDLINE | ID: mdl-11733576

ABSTRACT

Tumor necrosis factor (TNF) is a proinflammatory cytokine, which is centrally involved in several inflammatory disorders. Administration of TNF leads to a potentially lethal systemic inflammatory response syndrome (SIRS). We observed that (a) mice lacking functional genes for metallothionein 1 and 2 (MT-null) were protected compared with wild-type controls (P = 0.0078), and (b) mice overexpressing MT-1 (MT-TG) were more sensitized for the lethal effect of TNF than control mice (P = 0.0003), indicating a mediating role for MT in TNF induced SIRS. As MT is involved in the body zinc homeostasis, we tested whether zinc-deprivation or -supplementation alters the response to TNF. Although zinc-depletion strongly sensitized (P = 0.036), and pretreatment with zinc sulfate (ZnSO4) conferred protection against the deleterious effects of TNF (P < 0.0002), it was also found that the protection provided by zinc is independent of MT. Our observation that hsp70 is strongly induced in jejunum after ZnSO4 treatment, suggests a contribution of hsp70 in the protection against TNF. In addition, ZnSO4 cotreatment allowed complete regression of inoculated tumors with TNF and interferon gamma, leading to a significantly better survival (P = 0.0045).


Subject(s)
Dietary Supplements , Metallothionein/physiology , Systemic Inflammatory Response Syndrome/metabolism , Tumor Necrosis Factor-alpha/toxicity , Zinc/metabolism , Animals , Female , Melanoma, Experimental/metabolism , Metallothionein/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Systemic Inflammatory Response Syndrome/chemically induced , Systemic Inflammatory Response Syndrome/mortality , Tumor Necrosis Factor-alpha/administration & dosage
10.
Cytokine ; 15(2): 59-65, 2001 Jul 21.
Article in English | MEDLINE | ID: mdl-11500080

ABSTRACT

Tumour necrosis factor (TNF) is a major mediator in septic shock and several inflammatory diseases such as hepatitis. Galactosamine (GalN) sensitises experimental animals for TNF and the combination TNF/GalN leads to a lethal inflammatory hepatitis. We describe that a single injection of lipopolysaccharide (LPS), interleukin-1 (IL-1) or TNF can desensitise against the lethality induced by TNF/GalN, but also against changes in metabolic parameters such as hypothermia and transaminase release, in a dose responsive way. We also describe the desensitising capacity of a component present in Mouse Liver Extract (MLE). The MLE desensitises mice against the effects of TNF/GalN in a dose responsive way. The activity of the MLE is heat labile and does not involve LPS, TNF, IL-1 or TNF soluble receptors. We describe partial and complete purification of the factor. Partially pure material protects mice against all changes induced by TNF/GalN. The protection is dose dependent and heat labile and also possible in endotoxin-hyporesponsive C3H/HeJ mice. The pure material protects against lethality, hypothermia and AST release and it appears as a heat labile protein of relative molecular weight of 70 kDa probably with a break down product of 35 kDa.


Subject(s)
Galactosamine/antagonists & inhibitors , Interleukin-1/administration & dosage , Lipopolysaccharides/administration & dosage , Liver Extracts/isolation & purification , Liver Extracts/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Aspartate Aminotransferases/blood , Desensitization, Immunologic , Dose-Response Relationship, Drug , Female , Galactosamine/immunology , Galactosamine/toxicity , Injections, Intraperitoneal , Interleukin-1/immunology , Lipopolysaccharides/immunology , Liver/chemistry , Liver Extracts/chemistry , Mice , Mice, Inbred C57BL , Molecular Weight , Proteins/immunology , Proteins/isolation & purification , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/toxicity
11.
J Biol Chem ; 276(40): 37426-30, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11448951

ABSTRACT

The bioactivity of tumor necrosis factor (TNF) is mediated by two TNF receptors (TNF-Rs), more particularly TNF-RI and TNF-RII. Although human TNF (hTNF) and murine TNF (mTNF) are very homologous, hTNF binds only to mTNF-RI. By measuring the binding of a panel of mTNF/hTNF chimeras to both mTNF-R, we pinpointed the TNF region that mediates the interaction with mTNF-RII. Using site-specific mutagenesis, we identified amino acids 71-73 and 89 as the main interacting residues. Mutein hTNF-S71D/T72Y/H73 Delta/T89E interacts with both types of mTNF-R and is active in CT6 cell proliferation assays mediated by mTNF-RII. Mutein mTNF-D71S/Y72T/Delta 73H/E89T binds to mTNF-RI only and is no longer active on CT6 cells. However, the L929s cytotoxicity of this mutein (an effect mediated by mTNF-RI triggering) was also 100-fold lower than that of wild-type mTNF due to enhanced dissociation during incubation at subnanomolar concentrations. The additional mutation of amino acid 102, resulting in the mutein mTNF-D71S/Y72T/Delta 73H/E89T/P102Q, restored the trimer stability, which led to an enhanced specific activity on L929s cells. Hence the specific activity of a TNF species is governed not only by its receptor binding characteristics but also by its trimer stability after incubation at subnanomolar concentrations. In conclusion, the mutation of TNF amino acids 71-73, 89, and 102 is sufficient to obtain a mTNF mutein selective for mTNF-RI and a hTNF mutein that, unlike wild-type hTNF, also acts on mTNF-RII.


Subject(s)
Antigens, CD/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , Antigens, CD/genetics , Chromatography, Gel , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics
12.
J Biol Chem ; 276(29): 27098-103, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11371574

ABSTRACT

Incubation of murine tumor necrosis factor (mTNF) at subnanomolar concentrations results in partial dissociation of the trimers, coinciding with a decrease in bioactivity. Using size-exclusion chromatography, we observed that the conversion of labeled mTNF to monomers is not only prevented by coincubation with an excess of unlabeled mTNF but also with unlabeled human TNF (hTNF). Moreover, after coincubation of mTNF and hTNF four different TNF complexes were revealed by native polyacrylamide gel electrophoresis, viz. homotrimeric mTNF and hTNF, as well as two complexes with an intermediate migration pattern. Analytical gel filtration in combination with native polyacrylamide gel electrophoresis and Western blot immunodetection indicated that these new complexes consisted of heterotrimeric TNF molecules. We conclude that an exchange of monomers takes place during coincubation of two different species of TNF, which results in homotrimeric and heterotrimeric TNF. To assess receptor interaction in vitro, TNF heterotrimeric molecules were used as obtained after incubation of mTNF with labeled hTNF (which only binds to mTNF receptor I) or with labeled mutein mTNF75 (specific for mTNF receptor II). These heterotrimers were retained by both mTNF receptors, which means that the mTNF subunits incorporated in heterotrimeric complexes still can bind to both types of TNF receptor. In addition, the gradual decrease in mTNF bioactivity during preincubation at subnanomolar concentrations was prevented by the presence of mutein mTNF75, which is inactive in an L929 cytotoxicity assay, indicating that heterotrimerization can influence the overall bioactivity.


Subject(s)
Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies, Monoclonal/immunology , Biopolymers , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Humans , Mice , Species Specificity , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/isolation & purification
13.
Eur Cytokine Netw ; 11(4): 597-601, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11125302

ABSTRACT

Tumor necrosis factor (TNF) is an essential mediator in the pathogenesis of Gram-negative septic shock. Injection of TNF into normal mice leads to systemic, lethal inflammation, which is indistinguishable from lipopolysaccharide (LPS)-induced lethal inflammation. alpha(2)-macroglobulin (A2M) is a major positive acute phase protein with broad-spectrum protease-inhibitory activity. Mouse A2M-deficient (MAM-/-) mice were significantly protected against lethal systemic inflammation induced by TNF. The protection is not due to faster clearance of the injected TNF. The induction of tolerance to TNF-induced lethality by repetitive administration of small doses of human TNF for five consecutive days was equally efficient in both mutant mice compared to wild-type mice. In D-(+)-galactosamine (GalN)-sensitized mice, TNF induces lethal inflammatory hepatitis. MAM(-/-) mice are equally sensitive to the lethal combination of TNF/GalN. Furthermore, interleukin-1-induced desensitization to TNF/GalN was not impaired in MAM(-/-) mice. We conclude that MAM plays a mediating role in TNF-induced lethal shock and that MAM deficiency does not reduce changes in efficiency of tolerance and desensitization to TNF and TNF/GalN-induced lethality, respectively.


Subject(s)
Shock/chemically induced , Tumor Necrosis Factor-alpha/pharmacology , alpha-Macroglobulins/deficiency , Animals , Desensitization, Immunologic , Galactosamine/pharmacology , Hepatitis, Animal/chemically induced , Inflammation Mediators/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protease Inhibitors/immunology , alpha-Macroglobulins/genetics , alpha-Macroglobulins/immunology
14.
Immunity ; 13(2): 223-31, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10981965

ABSTRACT

Hypotension and shock observed in sepsis, SIRS, and tumor necrosis factor (TNF) or cytokine-based cancer treatment are the consequence of excessive nitric oxide (NO) production and subsequent soluble guanylate cyclase (sGC)-mediated vascular smooth muscle relaxation. We demonstrate here that, while NO synthase (NOS) inhibitors exacerbated toxicity, inhibitors of sGC activation protected against TNF-induced lethality, bradycardia, and hypotension. Importantly, sGC inhibition did not interfere with the antitumor activity of TNF. Using NOS inhibitors or iNOS-deficient animals, we furthermore observed that no protection against TNF toxicity could be obtained in the absence of NO. These data imply that iNOS- (and not eNOS-) derived NO is an endogenous protective molecule indispensable to survive a TNF challenge and exerting this beneficial effect via sGC-independent mechanisms.


Subject(s)
Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Shock/metabolism , Shock/prevention & control , Tumor Necrosis Factor-alpha/toxicity , Animals , Enzyme Inhibitors/therapeutic use , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II
15.
Infect Immun ; 68(9): 5026-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10948120

ABSTRACT

The proinflammatory cytokine tumor necrosis factor alpha (TNF-alpha) induces lethal hepatitis when injected into D-(+)-galactosamine-sensitized mice on the one hand or systemic inflammatory response syndrome (SIRS) in normal mice on the other hand. We studied whether serum amyloid P component (SAP), the major acute-phase protein in mice, plays a protective role in both lethal models. For this purpose, we used SAP(0/0) mice generated by gene targeting. We studied the lethal response of SAP(0/0) or SAP(+/+) mice to both lethal triggers but found no differences in the sensitivity of both types of mice. We also investigated whether SAP is involved in establishing two types of endogenous protection: one using a single injection of interleukin-1beta (IL-1beta) for desensitization and clearly involving a liver protein, the other by tolerizing mice for 5 days using small doses of human TNF-alpha. Although after IL-1beta or after tolerization the SAP levels in the serum had risen fourfold in the control mice and not in the SAP(0/0) mice, the same extents of desensitization and tolerization were achieved. Finally, we observed that the induction of hemorrhagic necrosis in the skin of mice by two consecutive local injections with TNF-alpha was not altered in SAP(0/0) mice. We conclude that the presence or absence of SAP has no influence on the sensitivity of mice to TNF-alpha-induced hepatitis, SIRS, and hemorrhagic necrosis or on the endogenous protective mechanisms of desensitization or tolerization.


Subject(s)
Hepatitis, Animal/etiology , Serum Amyloid P-Component/physiology , Skin/pathology , Systemic Inflammatory Response Syndrome/etiology , Tumor Necrosis Factor-alpha/toxicity , Animals , Galactosamine , Interleukin-1/pharmacology , Mice , Mice, Inbred C57BL , Necrosis
16.
FEBS Lett ; 467(2-3): 201-5, 2000 Feb 11.
Article in English | MEDLINE | ID: mdl-10675538

ABSTRACT

Tumor necrosis factor (TNF) induces hepatitis when injected in human beings or in rodents. The molecular mechanism by which TNF induces hepatic distress remains largely unknown, although induction of apoptosis of hepatocytes appears to be an essential step. In order to increase the therapeutic value of TNF, we have studied the protective activity of several molecules and found that four chemically totally different substances confer significant protection in the model of TNF-induced lethal hepatitis in mice sensitized with D-(+)-galactosamine (GalN), but not in mice sensitized with actinomycin-D (ActD) or against anti-Fas-induced lethal hepatitis. Verapamil, a calcium-channel blocker, tannic acid, picotamide, a thromboxane A(2) receptor antagonist, and K76COOH, an inhibitor, amongst others, of complement, protected significantly against induction of lethality, release of the liver-specific enzyme alanine aminotransferase (ALT) and induction of apoptosis in the liver after TNF/GalN, except for K76COOH, which paradoxically increased ALT values after challenge, and which also protected against TNF/GalN in complement-deficient mice. The data suggest that activation of platelets and neutrophils, as well as induction of inflammation occur in the TNF/GalN model, but not in the TNF/ActD or anti-Fas models, in which direct induction of apoptosis of hepatocytes may be more relevant. The protective activity of the drugs may lead to an increase in therapeutic value of TNF.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Hydrolyzable Tannins/pharmacology , Phthalic Acids/pharmacology , Sesquiterpenes/pharmacology , Tumor Necrosis Factor-alpha/toxicity , Verapamil/pharmacology , Alanine Transaminase/blood , Animals , Apoptosis , Astringents/pharmacology , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Complement Inactivator Proteins/pharmacology , Dactinomycin/administration & dosage , Disease Models, Animal , Galactosamine/administration & dosage , Liver/drug effects , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Platelet Aggregation Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/administration & dosage
17.
J Immunol ; 163(10): 5235-41, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10553044

ABSTRACT

Lethal hepatitis can be induced by an agonistic anti-Fas Ab in normal mice or by TNF in mice sensitized to d -(+)-galactosamine or actinomycin D. In all three models, we found that apoptosis of hepatocytes is an early and necessary step to cause lethality. In the three models, we observed activation of the major executioner caspases-3 and -7. Two acute-phase proteins, alpha1-acid glycoprotein and alpha1-antitrypsin, differentially prevent lethality: alpha1-acid glycoprotein protects in both TNF models and not in the anti-Fas model, while alpha1-antitrypsin confers protection in the TNF/d -(+)-galactosamine model only. The protection is inversely correlated with activation of caspase-3 and caspase-7. The data suggest that activation of caspase-3 and -7 is essential in the in vivo induction of apoptosis leading to lethal hepatitis and that acute phase proteins are powerful inhibitors of apoptosis and caspase activation. Furthermore, Bcl-2 transgenic mice, expressing Bcl-2 specifically in hepatocytes, are protected against a lethal challenge with anti-Fas or with TNF/d -(+)-galactosamine, but not against TNF/actinomycin D. The acute-phase proteins might constitute an inducible anti-apoptotic protective system, which in pathology or disturbed homeostasis prevents excessive apoptosis.


Subject(s)
Acute-Phase Proteins/pharmacology , Caspase Inhibitors , Caspases/metabolism , Hepatitis, Animal/enzymology , Hepatitis, Animal/prevention & control , Alanine Transaminase/blood , Animals , Apoptosis , Enzyme Activation , Female , Hepatitis, Animal/mortality , Hepatitis, Animal/pathology , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Orosomucoid/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Necrosis Factor-alpha/pharmacology , alpha 1-Antitrypsin/pharmacology
18.
Cytokine ; 11(8): 617-25, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433810

ABSTRACT

Injection of tumour necrosis factor (TNF) in animals causes severe liver cell toxicity, especially when D-(+)-galactosamine (GalN) is co-administered. After challenge with TNF/GalN, serum complement activity (CH50 and APCH50) decreased dramatically, suggesting strong activation of both the classical and the alternative pathways. TNF or GalN alone had no such effect. A cleavage product of complement protein C3 [C3(b)] was deposited on the surface of hepatocytes of TNF/GalN-treated mice. Intravenous administration of cobra venom factor (CVF), which depletes complement, inhibited the development of hepatitis. However, CVF pretreatment also protected C3-deficient mice. Pretreatment of mice with a C1q-depleting antibody did not prevent TNF/GalN lethality, although the anti-C1q antibody had depleted plasma C1q. Factor B-deficient and C3-deficient mice, generated by gene targeting, proved to be as sensitive to TNF/GalN as control mice. Furthermore, induction of lethal shock by platelet-activating factor, an important mediator in TNF-induced hepatic failure, was not reduced in C3-deficient mice. These data indicate that complement, although activated, plays no major role in the generation of acute lethal hepatic failure in this model and that CVF-induced protection is independent of complement depletion.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Complement Activation/immunology , Liver/immunology , Tumor Necrosis Factor-alpha/toxicity , Animals , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Complement Activation/drug effects , Complement C1q/analysis , Complement C3b/analysis , Complement Pathway, Alternative/immunology , Complement Pathway, Classical/immunology , Elapid Venoms/pharmacology , Female , Galactosamine/toxicity , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA
19.
FEBS Lett ; 445(1): 115-8, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-10069384

ABSTRACT

Experimental hepatitis induced by tumor necrosis factor in D-(+)-galactosamine-sensitized mice or by an agonistic anti-Fas antibody in normal mice is accompanied by dramatic apoptosis of hepatocytes. Apoptosis is the final result of activation of a cascade of caspases. We used caspase-1-/- mice, generated by gene targeting, to study the role of this protease in TNF- and anti-Fas-induced lethal hepatitis. We found that mutant mice exhibited the typical caspase-1-/- phenotype, since they resisted to a lethal injection of LPS and released no interleukin-1beta in the circulation, in contrast to wild-type littermates. When caspase-1-/- mice were challenged with different doses of tumor necrosis factor/D-(+)-galactosamine or with anti-Fas, no increased survival was observed compared with control mice. Furthermore, apoptosis in the livers of these mice and serum levels of alanine aminotransferase were not reduced. These data indicate that caspase-1 deficiency does not lead to reduced apoptosis in these models, either because caspase-1 is irrelevant in this model or because of functional redundancy.


Subject(s)
Caspase 1/physiology , Hepatitis/etiology , Animals , Caspase 1/genetics , DNA Fragmentation , Disease Models, Animal , Female , Galactosamine/metabolism , Galactosamine/pharmacology , Interleukin-1/biosynthesis , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/immunology , fas Receptor/metabolism
20.
Genomics ; 55(3): 284-9, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-10049582

ABSTRACT

Administration of recombinant murine tumor necrosis factor (TNF) to mice results in lethal shock, characterized by hypotension, hypothermia, and dramatic induction of cytokines released in the circulation, such as interleukin-6 (IL-6). The sensitivity of mice to the effects of murine TNF varies from strain to strain. DBA/2 mice were found to be considerably more resistant to TNF than C57BL/6 mice. The resistance proved to be dominant since (C57BL/6 x DBA/2)F1 mice were also resistant. Using BXD recombinant inbred mice and a dose of TNF lethal for C57BL/6 but not for DBA/2 mice, we found that the resistance to TNF links to loci coding for corticosteroid-binding globulin (Cbg), alpha1-protease inhibitor (Spi1), contrapsin (Spi2) and the contrapsin-regulating gene Spi2r that form a gene cluster on chromosome 12. Quantitative trait-loci analysis of TNF-induced induction of IL-6 and of hypothermia also points to the importance of this locus (P < 0.0002 and P = 0.017, respectively), more particularly the Cbg and Spi2 loci, in the resistance to TNF. We propose to name the locus "TNF protection locus." The data suggest that endogenous protease inhibitors and/or glucocorticoids play a significant role in the attenuation of TNF-induced lethal shock. This study also demonstrates that loci affecting important biological responses can be identified with very high resolution using recombinant inbred mice.


Subject(s)
Tumor Necrosis Factor-alpha/toxicity , Animals , Dose-Response Relationship, Drug , Genotype , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Quantitative Trait, Heritable , Recombinant Proteins/pharmacology , Systemic Inflammatory Response Syndrome/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...