Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Am J Transplant ; 9(4): 675-86, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19298452

ABSTRACT

An important role of TNF interacting with TNFR2 has been shown in different models of ischemic, nephrotoxic and immune-mediated renal injury. To systematically evaluate the expression of TNFR2 in renal allograft rejection, we investigated human renal allograft biopsies and, in addition, established an experimental transplantation model in rats to verify the human data under standardized conditions. The expression of TNFR2 was analyzed in 96 human renal allograft biopsies with different disease entities. In a 6-day and a 28-day experimental protocol, TNFR2 was examined in kidney specimens and in the urine of control, uni-nephrectomized and transplanted rats +/- cyclosporine treatment (n = 114). In human biopsies and in rat allografts on day 6 with acute allograft rejection, significantly elevated expression of TNFR2 was observed in tubular epithelial cells, podocytes, B cells and monocytes/macrophages. The expression level was associated with renal function. The TNFR2 expression level at day 28 was significantly lower compared to day 6. TNFR2 is markedly upregulated both in human and experimental acute renal allograft rejection. Our data are robust and consistent between different species, suggesting a role for TNFR2 in the early course of rejection.


Subject(s)
Gene Expression Regulation , Graft Rejection/genetics , Kidney Transplantation/pathology , Receptors, Tumor Necrosis Factor, Type II/genetics , Adult , Aged , Animals , Biopsy , Cyclosporine/therapeutic use , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Models, Animal , Rats , Receptors, Tumor Necrosis Factor, Type I/physiology , Transplantation, Homologous/pathology , Up-Regulation
2.
Int J Immunopathol Pharmacol ; 21(3): 577-84, 2008.
Article in English | MEDLINE | ID: mdl-18831924

ABSTRACT

In contrast to humans, mice physiologically exhibit extramedullary haematopoiesis in the spleen. In spite of this crucial species specific difference not much is known about the contribution of extramedullary haematopoiesis to overall erythropoiesis in models of anaemia of inflammation (AI). The objective of this study is to characterize murine AI with respect to extramedullary haematopoiesis and to develop a model more closely resembling human AI. Three different models of AI [caecal ligation and puncture (CLP), collagen induced arthritis (CIA) and DSS induced chronic colitis (DSSC)] were characterized with respect to red blood parameters, iron metabolism and extramedullary haematopoiesis. Arthritic animals were splenectomised to prevent extramedullary haematopoiesis. Anaemia caused by systemic inflammation was found in all three models. Splenic extramedullary haematopoiesis was markedly increased as reflected by increment in spleen weights and increase of the red pulp resulting in increased reticulocyte counts. Splenectomised arthritic animals did not show increased reticulocyte counts indicating that most of the reticulocytes were produced in the spleen. Our results demonstrate that murine AI differs from human AI with respect to increased splenic extramedullary haematopoiesis. Our data demonstrate that induction of AI in splenectomised mice represents a good way to model human AI.


Subject(s)
Anemia/etiology , Hematopoiesis, Extramedullary , Inflammation/complications , Splenectomy , Anemia/therapy , Animals , Arthritis, Experimental/complications , Colitis/complications , Disease Models, Animal , Erythropoiesis , Female , Humans , Male , Mice , Mice, Inbred DBA , Species Specificity
3.
Infect Immun ; 73(10): 6488-92, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16177321

ABSTRACT

Nosocomial infections in immune-suppressed patients are a widespread problem in intensive care medicine. Such patients are highly susceptible to infections because their immune defenses are impaired and, therefore, unable to adequately combat invading microorganisms. To investigate the problem of sepsis-induced immune suppression, we used a model in which mice developed sublethal peritonitis induced by cecal ligation and puncture (CLP). Two days after CLP mice were in an immune-suppressed state, as measured by impaired capacity to produce tumor necrosis factor (TNF) and enhanced susceptibility to bacterial infections. Since macrophage migration inhibitory factor (MIF) is a critical mediator of septic shock by modulation of innate immune responses, the role of MIF in sepsis-induced immune suppression was analyzed. Neutralization of endogenous MIF further enhanced susceptibility to bacterial superinfection after CLP. Conversely, treatment with recombinant human MIF before the bacterial superinfection protected the animals. MIF treatment reconstituted the impaired capacity to produce proinflammatory cytokines, such as TNF and interleukin-6. This study indicates that MIF might be able to ameliorate the sepsis-induced immune suppression by reenabling the organism to react adequately to a secondary bacterial challenge.


Subject(s)
Bacterial Infections/immunology , Bacterial Infections/prevention & control , Immune Tolerance , Macrophage Migration-Inhibitory Factors/therapeutic use , Superinfection/immunology , Superinfection/prevention & control , Animals , Cytokines/metabolism , Female , Immunity, Innate/drug effects , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/pharmacology , Mice , Shock, Septic/immunology , Shock, Septic/metabolism , Tumor Necrosis Factors/metabolism
4.
Clin Exp Immunol ; 136(1): 21-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15030510

ABSTRACT

The lymphotoxin-beta receptor (LTbetaR) pathway is critical for maintenance of organized lymphoid structures and is involved in the development of colitis. To investigate the mechanisms by which LTbetaR activation contributes to the pathology of chronic inflammation we used a soluble LTbetaR-Ig fusion protein as a competitive inhibitor of LTbetaR activation in the mouse model of chronic colitis induced by oral administration of dextran sulphate sodium. Strong expression of LTbeta which constitutes part of the LTalpha(1)beta(2) ligand complex was detected in colonic tissue of mice with chronic colitis. Treatment with LTbetaR-Ig significantly attenuated the development and histological manifestations of the chronic inflammation and reduced the production of inflammatory cytokines such as TNF, IL-1beta, and IL-6. Moreover, LTbetaR-Ig treatment significantly down-regulated mucosal addressin cell adhesion molecule-1 (MAdCAM-1) expression, leading to reduced leucocyte rolling and sticking in postcapillary and collecting venules and reduced extravasation into the intestinal mucosa as quantified by in vivo fluorescence microscopy. Thus, LTbetaR pathway inhibition ameliorates DSS-induced experimental chronic colitis in mice by MAdCAM-1 down-regulation entailing reduced lymphocyte margination and extravasation into the inflamed mucosa. Therefore, a combined treatment with reagents blocking T cell-mediated perpetuation of chronic inflammation such as LTbetaR-Ig together with direct anti-inflammatory reagents such as TNF inhibitors could constitute a promising treatment strategy for chronic colitis.


Subject(s)
Colitis, Ulcerative/prevention & control , Immunoglobulins/metabolism , Mucoproteins/metabolism , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Animals , Cell Adhesion Molecules , Chronic Disease , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Down-Regulation/drug effects , Female , Intestinal Mucosa/blood supply , Intestinal Mucosa/metabolism , Leukocytes/physiology , Ligands , Lymphotoxin beta Receptor , Mice , Mice, Inbred BALB C , Microcirculation , Receptors, Tumor Necrosis Factor/physiology
5.
Infect Immun ; 69(12): 7271-6, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11705897

ABSTRACT

Loss, reduction, or enhancement of the ability to respond to bacterial lipopolysaccharide (LPS) has no influence on survival of mice in a model of postoperative polymicrobial septic peritonitis induced by cecal ligation and puncture (CLP). This was demonstrated by using either mice with a defective Tlr4 gene, which encodes the critical receptor molecule for LPS responses, or mice deficient for LPS binding protein (LBP) or mice sensitized to LPS by Propionibacterium acnes. Though interleukin-12 (IL-12) and gamma interferon (IFN-gamma) play an important role in the sensitivity to LPS as well as in the resistance to several infections, loss of these cytokine pathways does not affect survival after CLP. Thus, neutralization of neither endogenous IL-12 nor IFN-gamma altered mortality. In addition, IFN-gamma receptor-deficient mice demonstrated the same sensitivity to CLP as mice with a functional IFN-gamma receptor. However, administration of IFN-gamma at the time of operation or pretreatment of both IFN-gamma-sensitive and IFN-gamma-resistant mice with IL-12 significantly enhanced mortality. This indicates that in the present infection model activation of innate defense mechanisms is not dependent on LPS recognition and does not require endogenous IL-12 or IFN-gamma function. Indeed, exogenous application of these two mediators had deleterious effects.


Subject(s)
Acute-Phase Proteins , Bacterial Infections/immunology , Drosophila Proteins , Endotoxemia/immunology , Peritonitis/immunology , Postoperative Complications , Sepsis/immunology , Animals , Carrier Proteins/genetics , Cecum/surgery , Endotoxemia/mortality , Interferon-gamma/immunology , Interleukin-12/immunology , Ligation , Lipopolysaccharides/immunology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Peritonitis/mortality , Receptors, Cell Surface/genetics , Sepsis/mortality , Toll-Like Receptor 4 , Toll-Like Receptors
6.
J Interferon Cytokine Res ; 21(9): 757-62, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11576469

ABSTRACT

Tumor necrosis factor (TNF) exerts its biologic activity via two distinct membrane receptors, TNF receptor type 1 (p55TNFR) and TNF receptor type 2 (p75TNFR). Whereas the p55TNFR gene is rather constitutively expressed, transcription of p75TNFR is strongly modulated by a number of stimulatory agents. Experimental evidence suggested the involvement of p75TNFR in endothelial cell activation. Therefore, we have tested the transcriptional activity of p75TNFR under conditions of hypoxia and reoxygenation. Northern blot analysis revealed that p75TNFR mRNA is upregulated in NIH3T3 cells under hypoxia and reoxygenation. This observation directly originates from transcriptional activation of the p75TNFR gene, as shown by reporter gene analysis. Cotransfection experiments clearly showed that the transcriptional induction of the p75TNFR gene is independent of the hypoxia-induced factors, HIF-1alpha and HIF-2alpha. Using deletion mutants of the 5'-flanking region of the p75TNFR gene, we were able to identify a putative DNA binding site for the transcription factor nuclear factor-interleukin-6 (NF-IL-6) to be responsible for the transcriptional upregulation of the p75TNFR gene under conditions of hypoxia and reoxygenation.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Hypoxia/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Transcription Factors , Transcriptional Activation/genetics , 3T3 Cells/metabolism , Animals , Binding Sites/genetics , Binding Sites/physiology , DNA-Binding Proteins/metabolism , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Nuclear Proteins/metabolism , Peptide Initiation Factors/metabolism , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Type II , Up-Regulation/physiology
7.
Cancer Res ; 61(14): 5523-8, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454702

ABSTRACT

The mechanisms involved in the antimetastatic effect of CpG-containing DNA were investigated in a mouse model of experimental metastasis. Tumor cell colony formation in lungs or livers of mice after i.v. inoculation with syngeneic fibrosarcoma or thymoma cells was determined. The i.v. injection of plasmid DNA or synthetic oligodeoxynucleotides (ODNs) containing unmethylated CpG motifs before tumor cell application strongly inhibited metastasis. Because synthetic CpG-ODN was not directly tumor cytotoxic, the target cells for this CpG-ODN effect were determined. The cytotoxic activity on standard natural killer (NK) targets as well as on fibrosarcoma cells of splenic NK cells and NKT cell-containing liver mononuclear cells derived from CpG-ODN-treated mice was strongly enhanced. Participation of NK/NKT cells in the CpG-induced antimetastatic effect was demonstrated by reduction of the antimetastatic effect in mice depleted of NK/NKT cells and beta2-microglobulin-deficient mice. Neutralization of interleukin 12, interleukin 18, or IFN-gamma did not interfere with the CpG-induced antimetastatic effect. However, in sera of CpG-ODN-treated mice, high levels of IFN-alpha were detected, and in IFN-alpha/beta receptor-deficient mice, the CpG-ODN-induced antimetastatic effect was strongly reduced. These data indicate that CpG-ODNs activate NK/NKT cells for antimetastatic activity indirectly via IFN-alpha/beta receptor activation. The exploitation of the stimulatory activity of CpG-ODN for the innate immune system might be a useful strategy for antimetastatic therapy.


Subject(s)
CpG Islands/genetics , DNA/administration & dosage , Interferon Type I/physiology , Neoplasm Metastasis/prevention & control , Animals , Antibodies, Monoclonal/pharmacology , Cytokines/immunology , Cytokines/physiology , Cytotoxicity Tests, Immunologic , DNA/metabolism , DNA Methylation , Dose-Response Relationship, Drug , Female , Interferon Type I/immunology , Interferon-alpha/immunology , Interferon-alpha/physiology , Killer Cells, Natural/immunology , Killer Cells, Natural/physiology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Mice, SCID , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Tumor Cells, Cultured
8.
J Immunol ; 167(3): 1624-8, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11466385

ABSTRACT

Acute and chronic hyperinflammation are of major clinical concern, and many treatment strategies are therefore directed to inactivating parts of the inflammatory system. However, survival depends on responding quickly to pathogen attack, and since the adaptive immune system requires several days to adequately react, we rely initially on a range of innate defenses, many of which operate by activating parts of the inflammatory network. For example, LPS-binding protein (LBP) can transfer the LPS of Gram-negative bacteria to CD14 on the surface of macrophages, and this initiates an inflammatory reaction. However, the importance of this chain of events in infection is unclear. First, the innate system is redundant, and bacteria have many components that may serve as targets for it. Second, LBP can transfer LPS to other acceptors that do not induce inflammation. In this study, we show that innate defense against a lethal peritoneal infection with Salmonella requires a direct proinflammatory involvement of LBP, and that this is a major nonredundant function of LBP in this infection model. This emphasizes that blocking the LBP-initiated inflammatory cascade disables an essential defense pathway. Any anti-inflammatory protection that may be achieved must be balanced against the risks inherent in blinding the innate system to the presence of Gram-negative pathogens.


Subject(s)
Acute-Phase Proteins , Carrier Proteins/physiology , Lipopolysaccharides/metabolism , Membrane Glycoproteins , Peritonitis/pathology , Peritonitis/prevention & control , Salmonella Infections, Animal/pathology , Salmonella Infections, Animal/prevention & control , Animals , Carrier Proteins/administration & dosage , Carrier Proteins/genetics , Genetic Predisposition to Disease , Humans , Injections, Intraperitoneal , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Knockout , Peritoneum/microbiology , Peritoneum/pathology , Peritonitis/genetics , Peritonitis/immunology , Phenotype , Recombinant Proteins/administration & dosage , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/immunology , Salmonella typhimurium/growth & development , Salmonella typhimurium/immunology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/therapeutic use
9.
J Interferon Cytokine Res ; 21(5): 333-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11429164

ABSTRACT

The heterotrimeric lymphotoxin alpha(1)beta(2) (LTalpha(1)beta(2)) complex and LIGHT, a new member of the tumor necrosis factor (TNF) superfamily, have been identified as membrane-anchored ligands for the LTbeta receptor (LTbetaR), a member of the TNF receptor (TNFR) superfamily. Although some of the biologic activities of this receptor have been described using either soluble LTalpha(1)beta(2) as a ligand or agonistic monoclonal antibodies (mAb), very little is known about the signaling of LIGHT via the LTbetaR. To gain more insight into the biologic functions of LIGHT, we generated a recombinant soluble form of human LIGHT (rsHuLIGHT). We demonstrate here that this rsHuLIGHT is capable of binding to the LTbetaR. Interestingly, receptor-mediated ligand precipitation analysis revealed that rsHuLIGHT bound only to human LTbetaR but not to mouse LTbetaR, indicating a species-specific receptor ligand interaction. Activation of A375 human melanoma cells by rsHuLIGHT induced an increased secretion of interleukin-8 (IL-8). Furthermore, rsHuLIGHT caused growth arrest of A375 cells even in the absence of interferon-gamma (IFN-gamma).


Subject(s)
Growth Inhibitors/pharmacology , Interleukin-8/metabolism , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/pharmacology , Receptors, Tumor Necrosis Factor/physiology , Receptors, Virus/physiology , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , COS Cells , Growth Inhibitors/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Ligands , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Melanoma/prevention & control , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Protein Binding/genetics , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Member 14 , Receptors, Virus/biosynthesis , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Solubility , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor Ligand Superfamily Member 14 , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
10.
Eur Cytokine Netw ; 12(2): 325-30, 2001.
Article in English | MEDLINE | ID: mdl-11399522

ABSTRACT

The lymphotoxin beta-receptor (LT beta R), a member of the tumor necrosis factor (TNF) receptor family, plays a crucial role in lymphoid organogenesis by signaling through its functional ligand LT alpha(1)beta(2). While the receptor is expressed on a wide range of cell types e.g. fibroblasts and monocytes, the ligand is expressed only on activated T, B and NK cells. Remarkably, no cell type has been identified so far that expresses both the receptor and the ligand. In order to characterize the mouse LT beta R gene expression on a molecular level, we isolated about 1 kb of the 5' flanking region of the LT beta R gene. Primer extension analysis revealed one transcriptional start site located at - 60 upstream of the ATG-containing first exon. Northern blot analysis showed that the LT beta R is abundantly expressed in the mouse fibroblast cell line NIH 3T3, and to a lesser extent, in the mouse macrophage-like cell line RAW 264.7. To determine whether the 5' flanking region exerts functional promoter activity, we generated deletion mutants fused to the luciferase reporter gene. Transfection experiments using these reporter gene constructs showed that the isolated 5' flanking region is transcriptionally active in NIH 3T3 and RAW 264.7 cells, and determined a minimum length required for the transcriptional activity of the LT beta R promoter in these cells. Further sequence analysis of the isolated 5' flanking region identified a number of putative DNA-binding sites for transcription factors. Interestingly, incubation of NIH 3T3 cells with dexamethasone resulted in an elevated mRNA level of the LT beta R gene. This effect was abolished by using the specific glucocorticoid receptor inhibitor RU486, indicating an increased transcriptional activity of the LT beta R promoter after glucocorticoid stimulation.


Subject(s)
Promoter Regions, Genetic , Receptors, Tumor Necrosis Factor/genetics , 3T3 Cells , Animals , Base Sequence , DNA , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Luciferases/genetics , Lymphotoxin beta Receptor , Mice , Molecular Sequence Data , Transcription, Genetic/drug effects , Transfection
11.
Infect Immun ; 69(6): 3550-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11349012

ABSTRACT

The occurrence of peritoneal adhesions in surgical patients is positively correlated with tumor necrosis factor (TNF) levels. In a model of septic peritonitis-cecal ligation and puncture-TNF neutralization prevented formation of peritoneal adhesions and increased mortality, most likely because localization of the septic focus was prevented. To discriminate between the coagulation-independent protective TNF effect and a potential protective procoagulant TNF effect, formation of peritoneal adhesions after CLP was inhibited with heparin, hirudin, or urokinase. Each treatment increased mortality and increased the number of bacteria in the peritoneal lavage fluid, kidney, and liver to various degrees. Under these experimental conditions, antibiotics prevented death. In coagulation-compromised mice, lethality was further enhanced by additional TNF neutralization. These findings demonstrate that peritoneal adhesions early in septic peritonitis are an important mechanism of innate immunity that prevents increased spread of bacteria and reduces mortality.


Subject(s)
Peritonitis/immunology , Sepsis/immunology , Tissue Adhesions/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cecum/pathology , Cecum/physiology , Colony Count, Microbial , Disease Models, Animal , Heparin/pharmacology , Hirudins/pharmacology , Immunity, Innate , Ligation , Male , Mice , Peritonitis/pathology , Sepsis/pathology , Tissue Adhesions/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Urokinase-Type Plasminogen Activator/pharmacology
12.
Eur Cytokine Netw ; 12(1): 45-50, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11282545

ABSTRACT

We investigated the role of the TNF receptors, type I (p55TNFR) and type II (p75TNFR), in a mouse model of contact hypersensitivity, i.e., a model of a delayed type hypersensitivity (DTH) allergic reaction. Mice deficient for p55TNFR or p75TNFR were used to investigate the functions of these receptors in development of the DTH reaction. We show that both TNF receptors have a strong influence on the overall outcome of the DTH reaction, with the two TNF receptors exerting distinct functions. Dendritic cells of mice lacking p55TNFR had a defect in allergen uptake but showed normal migration into regional lymph nodes. In contrast, dendritic cells of p75TNFR-deficient mice showed diminished migration into regional lymph nodes after allergen contact, whereas the allergen uptake was independent of the p75TNFR. Thus, both TNF receptors are required for the development of a complete DTH reaction.


Subject(s)
Antigens, CD/metabolism , Dermatitis, Allergic Contact/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Animals , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II
13.
Int J Cancer ; 91(4): 543-9, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11251979

ABSTRACT

In this study, we characterised the anti-tumour as well as the pro-metastatic activities of TNF mutants deficient in their lectin-like activity.1619 We report that, despite reduced systemic toxicity as compared to wild-type (wt) mTNF, a (T104A) and a (T104A-E106A-E109A) mTNF mutant (triple mTNF) retained most of their necrotic and tumouristatic activities, as measured in a CFS-1 fibrosarcoma and a B16BL6 melanoma tumour model, respectively. These mutants also conserved their anti-angiogenic activity, as measured in an in vitro endothelial morphogenesis assay.26 In contrast, the pro-metastatic activity of the T104A and the triple mTNF mutants in the CFS-1 fibrosarcoma and the 3LL-R Lewis lung carcinoma tumour model was significantly lower than that of the wt molecule. These results thus indicate that the lectin-like domain of TNF is not implicated in its necrotic, tumouristatic and anti-angiogenic activities, but that it can contribute to the pro-metastatic effect of the cytokine. In conclusion, in view of their reduced systemic toxicity and pro-metastatic capacity, but their retained anti-tumour activities, lectin-deficient TNF mutants might prove to be therapeutically interesting alternatives to wt TNF.


Subject(s)
Lectins/metabolism , Mutation , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics , Animals , Carcinoma, Lewis Lung , Cattle , Cell Adhesion , Collagen/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Lung/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Necrosis , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms, Experimental , Neovascularization, Pathologic , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/therapeutic use
14.
J Biol Chem ; 276(22): 19390-5, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11279196

ABSTRACT

We report the identification of a novel p75TNF receptor isoform termed icp75TNFR, which is generated by the use of an alternative transcriptional start site within the p75TNFR gene and characterized by regulated intracellular expression. The icp75TNFR protein has an apparent molecular mass of approximately 50 kDa and is recognized by antibodies generated against the transmembrane form of p75TNFR. The icp75TNFR binds the tumor necrosis factor(TNF) and mediates intracellular signaling. Overexpression of the icp75TNFR cDNA results in TNF-induced activation of NFkappaB in a TNF receptor-associated factor 2 (TRAF2)-dependent manner. Thus, our results provide an example for intracellular cytokine receptor activation.


Subject(s)
Antigens, CD/chemistry , NF-kappa B/metabolism , Protein Isoforms , Receptors, Tumor Necrosis Factor/chemistry , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cell Line , DNA, Complementary/metabolism , Enzyme Activation , Exons , Genes, Reporter , HeLa Cells , Humans , Mice , Microscopy, Fluorescence , Models, Genetic , Molecular Sequence Data , Open Reading Frames , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Tumor Necrosis Factor, Type II , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction , TNF Receptor-Associated Factor 2 , Transcription, Genetic , Transfection , Tumor Cells, Cultured , U937 Cells
15.
FASEB J ; 14(10): 1380-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10877831

ABSTRACT

It is believed that an inflammation-induced activation of the CNS leads to an inhibition of overshooting immune responses to prevent extensive local cytokine secretion. However, immunosuppression by the sympathetic nervous system may be unfavorable when bacteria are present locally and when TNF-alpha is necessary to overcome infection. We now report in a superfusion model, using mouse spleen slices, that although local Pseudomonas aeruginosa increased splenic TNF-alpha and IL-6 secretion severalfold over basal levels, electrically released neurotransmitters attenuated cytokine secretion to similar basal level as under bacteria-free conditions. Bacteria reversed noradrenergic inhibitory effector mechanisms: Under bacteria-free conditions, TNF-alpha secretion was very low and IL-6 secretion was mainly inhibited by alpha2-adrenoreceptor ligation. In the presence of bacteria, TNF-alpha and IL-6 secretion were high and IL-6 secretion was mainly inhibited by beta-adrenoreceptor ligation. The alpha- to beta-adrenoswitch of IL-6 inhibition in the presence of bacteria was mediated by the prior adrenergic regulation of TNF-alpha. In vivo, chemical abrogation of sympathetic inhibition reduced accumulation of bacteria in the spleen, which depended at least in part on TNF-alpha. This suggests that activation of the sympathetic nervous system may be a forerunner for accumulation of bacteria in tissue and consecutively sepsis due to intensified inhibition of TNF-alpha secretion.


Subject(s)
Interleukin-6/metabolism , Spleen/immunology , Sympathetic Nervous System/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Denervation , Electric Stimulation , Female , In Vitro Techniques , Inflammation/immunology , Mice , Models, Biological , Norepinephrine/physiology , Perfusion , Pseudomonas Infections/immunology , Pseudomonas Infections/physiopathology , Pseudomonas aeruginosa/pathogenicity , Receptors, Adrenergic, alpha/immunology , Receptors, Adrenergic, beta/immunology , Spleen/innervation , Spleen/physiopathology
16.
J Interferon Cytokine Res ; 20(5): 511-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10841080

ABSTRACT

Vascular endothelial growth factor (VEGF) plays an important role in the production of ascitic fluid associated with malignant tumor growth. In an experimental model for malignant ascites formation, mice were inoculated intraperitoneally with syngeneic mouse sarcoma tumor cells. Ascites development was not prevented by administering tumor necrosis factor (TNF) simultaneously with the tumor cell inoculation. When the malignant ascites was first drained and renewal of ascites was monitored, however, a TNF dose-dependent inhibition of ascitic fluid accumulation was observed. Northern blot analyses indicated transient downregulation by TNF on the expression of VEGF mRNA in tumor cells. Monoclonal antibody, (mAb) DC101 generated against the mouse VEGF receptor Flk-1 prevented the recurrence of malignant ascites in mice similar to TNF inhibition. In addition, exogenous soluble human Flt-1 used as an inhibitor of endogenous VEGF binding also inhibited ascites recurrence. These data demonstrate that the observed inhibitory effect of TNF on reestablishment of malignant ascites can be achieved equally by inhibition of the interaction of VEGF with its receptor Flk-1.


Subject(s)
Ascites/etiology , Ascites/metabolism , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Ascites/prevention & control , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/genetics , Female , Humans , Lymphokines/antagonists & inhibitors , Lymphokines/genetics , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor , Recurrence , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Am J Pathol ; 156(4): 1171-6, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10751341

ABSTRACT

Activation of endothelial cells, fibrin deposition, and coagulation within the tumor vasculature has been shown in vivo to correlate with the occurrence of tumor necrosis factor (TNF)-induced tumor necrosis in mice. In the present study we investigated which target cells mediate the TNF-induced necrosis in fibrosarcomas grown in wild type (wt), TNF receptor type 1-deficient (TNFRp55-/-), and TNF receptor type 2-deficient (TNFRp75-/-) mice. TNF administration resulted in tumor necrosis exclusively in wt and TNFRp75-/-, but not in TNFRp55-/- mice, indicating a dependence of TNF-mediated tumor necrosis on the expression of TNF receptor type 1. However, using wt and TNFRp55-/- fibrosarcomas in wt mice, we found that TNF-mediated tumor necrosis was completely independent of TNF receptor type 1 expression in tumor cells. Thus we could exclude any direct tumoricidal effect of TNF in this model. Soluble TNF induced leukostasis in wt and TNFRp75-/- mice but not in TNFRp55-/- mice. TNF-induced leukostasis in TNFRp55-/- mice was restored by adoptive bone marrow transplantation of wt hematopoietic cells, but TNF failed to induce tumor necrosis in these chimeric mice. Because TNF administration resulted in both activation and focal damage of tumor endothelium, TNF receptor type 1-expressing cells of the tumor vasculature, likely to be endothelial cells, appear to be target cells for mediating TNF-induced tumor necrosis.


Subject(s)
Endothelium, Vascular/metabolism , Fibrosarcoma/blood supply , Fibrosarcoma/pathology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Bone Marrow Transplantation , Chimera , Endothelium, Vascular/pathology , Female , Leukostasis/chemically induced , Leukostasis/surgery , Mice , Mice, Inbred C57BL/genetics , Necrosis , Neoplasm Transplantation , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
18.
Nat Med ; 6(2): 164-70, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10655104

ABSTRACT

Identification of new therapeutic targets for the management of septic shock remains imperative as all investigational therapies, including anti-tumor necrosis factor (TNF) and anti-interleukin (IL)-1 agents, have uniformly failed to lower the mortality of critically ill patients with severe sepsis. We report here that macrophage migration inhibitory factor (MIF) is a critical mediator of septic shock. High concentrations of MIF were detected in the peritoneal exudate fluid and in the systemic circulation of mice with bacterial peritonitis. Experiments performed in TNFalpha knockout mice allowed a direct evaluation of the part played by MIF in sepsis in the absence of this pivotal cytokine of inflammation. Anti-MIF antibody protected TNFalpha knockout from lethal peritonitis induced by cecal ligation and puncture (CLP), providing evidence of an intrinsic contribution of MIF to the pathogenesis of sepsis. Anti-MIF antibody also protected normal mice from lethal peritonitis induced by both CLP and Escherichia coli, even when treatment was started up to 8 hours after CLP. Conversely, co-injection of recombinant MIF and E. coli markedly increased the lethality of peritonitis. Finally, high concentrations of MIF were detected in the plasma of patients with severe sepsis or septic shock. These studies define a critical part for MIF in the pathogenesis of septic shock and identify a new target for therapeutic intervention.


Subject(s)
Bacterial Infections/prevention & control , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Shock, Septic/prevention & control , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Bacterial Infections/metabolism , Female , Humans , Macrophage Migration-Inhibitory Factors/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Male , Mice , Middle Aged , Peritonitis/metabolism , Peritonitis/prevention & control , Shock, Septic/metabolism
20.
Eur Cytokine Netw ; 10(4): 541-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10586121

ABSTRACT

Mechanisms involved in the antimetastatic effect of IL-12 were analyzed in a mouse model of experimental metastasis with either syngeneic fibrosarcoma cells colonizing the lungs or syngeneic B cell lymphoma cells colonizing the liver. IL-12 pretreatment effectively reduced the number of tumor colonies in both systems. This effect was already manifest 24 hours after tumor cell injection, indicating a T and B cell-independent mechanism. Therefore, the involvement of NK and alphabetaNKT cells was investigated using mice with defective NK and alphabetaNKT cell functions. Mice with impaired NK functions due to NK cell depletion, were less responsive to the antimetastatic IL-12 effect. IL-12 treatment failed to inhibit metastasis in beta2-microglobulin-deficient mice which lack alphabetaNKT cells in addition to having impaired NK cell activity, thus, demonstrating the functional importance of IL-12-activated NK and alphabetaNKT cells. While the IL-12-induced antimetastatic effect of NK cells was dependent on IFN-gamma action, IL-12 activation of alphabetaNKT cells did not involve IFN-gamma. The neutralization of IFN-gamma or the use of IFN-gamma receptor-deficient mice did not alter the IL-12-induced effect in the absence of NK cells. Activation of effector cells of the innate immune system, such as NK and alphabetaNKT cells, seems to be the main mechanism for the antimetastatic effect of IL-12.


Subject(s)
Interferon-gamma/immunology , Interleukin-12/therapeutic use , Killer Cells, Natural/immunology , Neoplasm Metastasis/drug therapy , T-Lymphocytes/immunology , Animals , Cytotoxicity, Immunologic/drug effects , Female , Fibrosarcoma/drug therapy , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Gene Deletion , Interferon-gamma/physiology , Interleukin-12/antagonists & inhibitors , Interleukin-12/immunology , Interleukin-12/pharmacology , Killer Cells, Natural/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymphoma/drug therapy , Lymphoma/immunology , Lymphoma/pathology , Mice , Mice, Inbred Strains , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Receptors, Interferon/genetics , Receptors, Interferon/physiology , Receptors, Interleukin-2/chemistry , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/physiology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , T-Lymphocytes/drug effects , Tumor Cells, Cultured , Tumor Stem Cell Assay , beta 2-Microglobulin/genetics , beta 2-Microglobulin/physiology , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...