Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 7(8): 1004-17, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25995337

ABSTRACT

TNF is a central actor during inflammation and a well-recognized drug target for inflammatory diseases. We found that the mouse strain SPRET/Ei, known for extreme and dominant resistance against TNF-induced shock, displays weak expression of TNF receptor 1 protein (TNFR1) but normal mRNA expression, a trait genetically linked to the major TNFR1 coding gene Tnfrsf1a and to a locus harbouring the predicted TNFR1-regulating miR-511. This miRNA is a genuine TNFR1 regulator in cells. In mice, overexpression of miR-511 down-regulates TNFR1 and protects against TNF, while anti-miR-511 up-regulates TNFR1 and sensitizes for TNF, breaking the resistance of SPRET/Ei. We found that miR-511 inhibits endotoxemia and experimental hepatitis and that this miR is strongly induced by glucocorticoids and is a true TNFR1 modulator and thus an anti-inflammatory miR. Since minimal reductions of TNFR1 have considerable effects on TNF sensitivity, we believe that at least part of the anti-inflammatory effects of glucocorti-coids are mediated by induction of this miR, resulting in reduced TNFR1 expression.


Subject(s)
Glucocorticoids/metabolism , MicroRNAs/metabolism , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Animals , Down-Regulation , Mice
2.
J Biol Chem ; 290(7): 4022-37, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25538244

ABSTRACT

The cytokine TNF is a well known drug target for several inflammatory diseases such as Crohn disease. Despite the great success of TNF blockers, therapy could be improved because of high costs and side effects. Selective inhibition of TNF receptor (TNFR) 1 signaling holds the potential to greatly reduce the pro-inflammatory activity of TNF, thereby preserving the advantageous immunomodulatory signals mediated by TNFR2. We generated a selective human TNFR1 inhibitor based on Nanobody (Nb) technology. Two anti-human TNFR1 Nbs were linked with an anti-albumin Nb to generate Nb Alb-70-96 named "TNF Receptor-One Silencer" (TROS). TROS selectively binds and inhibits TNF/TNFR1 and lymphotoxin-α/TNFR1 signaling with good affinity and IC50 values, both of which are in the nanomolar range. Surface plasmon resonance analysis reveals that TROS competes with TNF for binding to human TNFR1. In HEK293T cells, TROS strongly reduces TNF-induced gene expression, like IL8 and TNF, in a dose-dependent manner; and in ex vivo cultured colon biopsies of CD patients, TROS inhibits inflammation. Finally, in liver chimeric humanized mice, TROS antagonizes inflammation in a model of acute TNF-induced liver inflammation, reflected in reduced human IL8 expression in liver and reduced IL6 levels in serum. These results demonstrate the considerable potential of TROS and justify the evaluation of TROS in relevant disease animal models of both acute and chronic inflammation and eventually in patients.


Subject(s)
Colon/drug effects , Crohn Disease/prevention & control , Inflammation/prevention & control , Liver/drug effects , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/immunology , Single-Domain Antibodies/pharmacology , Amino Acid Sequence , Animals , Colon/immunology , Colon/pathology , Crohn Disease/immunology , Crohn Disease/pathology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Female , Humans , Inflammation/immunology , Inflammation/pathology , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Protein Conformation , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Surface Plasmon Resonance , Tumor Necrosis Factor-alpha/pharmacology
3.
Cytokine Growth Factor Rev ; 25(3): 285-300, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24746195

ABSTRACT

TNF is an essential regulator of the immune system. Dysregulation of TNF plays a role in the pathology of many auto-immune diseases. TNF-blocking agents have proven successful in the treatment of such diseases. Development of novel, safer or more effective drugs requires a deeper understanding of the regulation of the pro-inflammatory activities of TNF and its receptors. The ubiquitously expressed TNFR1 is responsible for most TNF effects, while TNFR2 has a limited expression pattern and performs immune-regulatory functions. Despite extensive knowledge of TNFR1 signaling, the regulation of TNFR1 expression, its modifications, localization and processing are less clear and the data are scattered. Here we review the current knowledge of TNFR1 regulation and discuss the impact this has on the host.


Subject(s)
Autoimmune Diseases/metabolism , Gene Expression Regulation , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Int J Cancer ; 135(3): 742-50, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24382818

ABSTRACT

Tumor necrosis factor (TNF) has remarkable antitumor effects, but its systemic therapeutic use is prevented by its lethal inflammatory effects. TNFR1 (P55) is essential for both the antitumor and toxic effects because both of them are absent in P55-deficient mice. In previous work we demonstrated that P55+/- mice are completely resistant to TNF toxicity, while the antitumor effects induced by TNF combined with interferon gamma (IFNγ) remain fully functional in these mice. Hence, a high dose of TNF/IFNγ has an excellent therapeutic potential when P55 levels are reduced, because TNF induces tumor regression without systemic toxicity. Here, we provide proof of principle for therapeutic application of this approach by using antisense oligonucleotides (ASOs). Treatment of mice with ASOs targeting P55 resulted in a strong reduction in P55 protein levels in liver, small intestine and blood mononuclear cells. This P55 downregulation was associated with significant protection of mice against acute TNF toxicity as measured by hypothermia, systemic inflammation and lethality. This treatment also protected mice against toxicity of TNF/IFNγ treatment in several cancer models: B16Bl6, Lewis lung carcinoma and a lung colony model. Our results confirm the therapeutic value of this strategy, which could lead to the development of a safer and more effective TNF/IFNγ antitumor therapy.


Subject(s)
Carcinoma, Lewis Lung/prevention & control , Disease Models, Animal , Interferon-gamma/toxicity , Melanoma, Experimental/prevention & control , Oligonucleotides, Antisense/pharmacology , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/toxicity , Animals , Carcinoma, Lewis Lung/chemically induced , Carcinoma, Lewis Lung/genetics , Female , Intestine, Small/metabolism , Liver/metabolism , Maximum Tolerated Dose , Melanoma, Experimental/chemically induced , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Signal Transduction
5.
J Clin Invest ; 123(6): 2590-603, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23676465

ABSTRACT

TNF has remarkable antitumor activities; however, therapeutic applications have not been possible because of the systemic and lethal proinflammatory effects induced by TNF. Both the antitumor and inflammatory effects of TNF are mediated by the TNF receptor p55 (p55TNFR) (encoded by the Tnfrsf1a gene). The antitumor effect stems from an induction of cell death in tumor endothelium, but the cell type that initiates the lethal inflammatory cascade has been unclear. Using conditional Tnfrsf1a knockout or reactivation mice, we found that the expression level of p55TNFR in intestinal epithelial cells (IECs) is a crucial determinant in TNF-induced lethal inflammation. Remarkably, tumor endothelium and IECs exhibited differential sensitivities to TNF when p55TNFR levels were reduced. Tumor-bearing Tnfrsf1a⁺⁺/⁻ or IEC-specific p55TNFR-deficient mice showed resistance to TNF-induced lethality, while the tumor endothelium remained fully responsive to TNF-induced apoptosis and tumors regressed. We demonstrate proof of principle for clinical application of this approach using neutralizing anti-human p55TNFR antibodies in human TNFRSF1A knockin mice. Our results uncover an important cellular basis of TNF toxicity and reveal that IEC-specific or systemic reduction of p55TNFR mitigates TNF toxicity without loss of antitumor efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Intestinal Mucosa/drug effects , Melanoma, Experimental/drug therapy , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis , Cytokines/genetics , Cytokines/metabolism , Drug Screening Assays, Antitumor , Endothelium/pathology , Gene Expression , Gene Knock-In Techniques , Humans , Inflammation/chemically induced , Interferon-gamma/physiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/therapeutic use , Tumor Necrosis Factor-alpha/toxicity
6.
EMBO Mol Med ; 5(3): 456-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23495141

ABSTRACT

Natural variation for LPS-induced lethal inflammation in mice is useful for identifying new genes that regulate sepsis, which could form the basis for novel therapies for systemic inflammation in humans. Here we report that LPS resistance of the inbred mouse strain SPRET/Ei, previously reported to depend on the glucocorticoid receptor (GR), maps to the distal region of the X-chromosome. The GR-inducible gene Tsc22d3, encoding the protein Gilz and located in the critical region on the X-chromosome, showed a higher expressed SPRET/Ei allele, regulated in cis. Higher Gilz levels were causally related to reduced inflammation, as shown with knockdown and overexpression studies in macrophages. Transient overexpression of Gilz by hydrodynamic plasmid injection confirmed that Gilz protects mice against endotoxemia Our data strongly suggest that Gilz is responsible for the LPS resistance of SPRET/Ei mice and that it could become a treatment option for sepsis.


Subject(s)
Endotoxemia/genetics , Inflammation/genetics , Lipopolysaccharides , Transcription Factors/genetics , X Chromosome , Animals , Cell Line , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/metabolism , Endotoxemia/prevention & control , Female , Gene Knockdown Techniques , Genetic Predisposition to Disease , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/prevention & control , Liver/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Orchiectomy , Ovariectomy , Phenotype , Quantitative Trait Loci , RNA Interference , Sex Factors , Time Factors , Transcription Factors/metabolism , Transfection
7.
J Neurosci ; 32(29): 9805-16, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22815495

ABSTRACT

Systemic inflammatory response syndrome (SIRS) is a highly mortal inflammatory disease, associated with systemic inflammation and organ dysfunction. SIRS can have a sterile cause or can be initiated by an infection, called sepsis. The prevalence is high, and available treatments are ineffective and mainly supportive. Consequently, there is an urgent need for new treatments. The brain is one of the first organs affected during SIRS, and sepsis and the consequent neurological complications, such as encephalopathy, are correlated with decreased survival. The choroid plexus (CP) that forms the blood-CSF barrier (BCSFB) is thought to act as a brain "immune sensor" involved in the communication between the peripheral immune system and the CNS. Nevertheless, the involvement of BCSFB integrity in systemic inflammatory diseases is seldom investigated. We report that matrix metalloprotease-8 (MMP8) depletion or inhibition protects mice from death and hypothermia in sepsis and renal ischemia/reperfusion. This effect could be attributed to MMP8-dependent leakage of the BCSFB, caused by collagen cleavage in the extracellular matrix of CP cells, which leads to a dramatic change in cellular morphology. Disruption of the BCSFB results in increased CSF cytokine levels, brain inflammation, and downregulation of the brain glucocorticoid receptor. This receptor is necessary for dampening the inflammatory response. Consequently, MMP8(+/+) mice, in contrast to MMP8(-/-) mice, show no anti-inflammatory response and this results in high mortality. In conclusion, we identify MMP8 as an essential mediator in SIRS and, hence, a potential drug target. We also propose that the mechanism of action of MMP8 involves disruption of the BCSFB integrity.


Subject(s)
Blood-Brain Barrier/metabolism , Endotoxemia/metabolism , Extracellular Matrix/metabolism , Matrix Metalloproteinase 8/metabolism , Systemic Inflammatory Response Syndrome/metabolism , Animals , Brain/metabolism , Choroid Plexus/metabolism , Collagen/metabolism , Corticosterone/blood , Female , Male , Mice , Mice, Knockout
9.
J Biol Chem ; 285(40): 31073-86, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20663891

ABSTRACT

SPRET/Ei mice are extremely resistant to acute LPS-induced lethal inflammation when compared with C57BL/6. We found that in vivo SPRET/Ei mice exhibit strongly reduced expression levels of cytokines and chemokines. To investigate the role of the potent anti-inflammatory glucocorticoid receptor (GR) in the SPRET/Ei phenotype, mice were treated with the GR antagonist RU486 or bilateral adrenalectomy. Under such conditions, both C57BL/6 and SPRET/Ei mice were strongly sensitized to LPS, and the differences in LPS response between SPRET/Ei and C57BL/6 mice were completely gone. These results underscore the central role of GR in the LPS hyporesponsiveness of SPRET/Ei mice. Compared with C57BL/6, SPRET/Ei mice were found to express higher GR levels, which were reflected in increased GR transactivation. Using a backcross mapping strategy, we demonstrate that the high GR transcription levels are linked to the Nr3c1 (GR) locus on chromosome 18 itself. Unexpectedly, SPRET/Ei mice exhibit a basal overactivation of the hypothalamic-pituitary-adrenal axis, namely strongly increased corticosterone levels, ACTH levels, and adrenocortical size. As a consequence of the excess of circulating glucocorticoids (GCs), levels of hepatic gluconeogenic enzymes are increased, and insulin secretion from pancreatic ß-cells is impaired, both of which result in hyperglycemia and glucose intolerance in SPRET/Ei mice. We conclude that SPRET/Ei mice are unique as they display an unusual combination of elevated GR expression and increased endogenous GC levels. Hence, these mice provide a new and powerful tool for the study of GR- and GC-mediated mechanisms, including immune repressive functions, neuroendocrine regulation, insulin secretion, and carbohydrate metabolism.


Subject(s)
Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Receptors, Glucocorticoid/biosynthesis , Adrenal Cortex/metabolism , Adrenalectomy , Adrenocorticotropic Hormone/metabolism , Animals , Chromosome Mapping , Chromosomes, Mammalian/genetics , Corticosterone/metabolism , Crosses, Genetic , Drug Resistance/drug effects , Drug Resistance/genetics , Gene Expression Regulation/genetics , Genetic Loci/genetics , Hormone Antagonists/pharmacology , Hypothalamo-Hypophyseal System/metabolism , Insulin-Secreting Cells/metabolism , Mice , Mifepristone/pharmacology , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/genetics
10.
J Immunol ; 178(8): 5069-75, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17404289

ABSTRACT

In our search for genes that inhibit the inflammatory effects of TNF without diminishing its antitumor capacities we found that, compared with C57BL/6 mice, DBA/2 mice exhibit a dominant resistance to TNF-induced lethality. Tumor-bearing (C57BL/6 x DBA/2)(BXD)F(1) mice completely survived an otherwise lethal TNF/IFN-gamma-antitumor therapy with complete regression of the tumor. This was not the case for C57BL/6 mice. Genetic linkage analysis revealed that TNF resistance is linked to a major locus on distal chromosome 6 and a minor locus on chromosome 17. Compared with littermate controls, chromosome substitution mice carrying a DBA/2 chromosome 6 in a C57BL/6 background were significantly protected against TNF and TNF/IFN-gamma, albeit less so than DBA/2 mice. Definition of a critical region of 13 Mb on chromosome 6 was the highest mapping resolution obtained. Further analysis of candidate genes may provide a powerful tool to control TNF-induced pathologies in humans.


Subject(s)
Chromosome Mapping , Shock/chemically induced , Tumor Necrosis Factor-alpha/toxicity , Animals , Female , Interferon-gamma/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Quantitative Trait Loci , Species Specificity , Tumor Necrosis Factor-alpha/administration & dosage
11.
J Immunol ; 175(11): 7642-9, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16301674

ABSTRACT

Acute fulminant liver failure is a serious worldwide health problem. Despite maximal supportive intensive care treatment, the disease offers a poor prognosis, with mortality rates of >80%. We have previously shown that a broad-spectrum inhibitor of matrix metalloproteinases (MMPs) confers complete protection in a mouse model of TNF-induced lethal hepatitis, thereby suggesting the possibility of protecting cancer patients against the deleterious side effects of TNF therapy. In our search for the individual matrix metalloproteinases involved, we found that the recently generated MMP-8-deficient mice are significantly protected against TNF-induced acute hepatitis. In contrast to their wild-type counterparts, MMP-8-null mice display very little hepatocyte necrosis and apoptosis, resulting in a much better survival outcome. We found that these animals clearly display impaired leukocyte influx into the liver and no release of the neutrophil-specific, LPS-induced CXC chemokine. Our findings provide evidence that MMP-8 plays an essential role in acute liver failure and might be a promising new target for the treatment for this illness.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Matrix Metalloproteinase 8/deficiency , Tumor Necrosis Factor-alpha/toxicity , Animals , Blotting, Western , Chemical and Drug Induced Liver Injury/pathology , Chemokine CXCL5 , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Disease Models, Animal , Immunohistochemistry , Liver/immunology , Liver/metabolism , Liver/pathology , Matrix Metalloproteinase 8/immunology , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Knockout , Neutrophil Infiltration/immunology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
12.
Mamm Genome ; 15(7): 537-43, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15366373

ABSTRACT

Most inflammatory disorders are becoming more prevalent, especially in Western countries. The pro-inflammatory cytokine tumor necrosis factor-alpha (TNF) plays a prominent role in many of these inflammatory disorders. We have previously shown that SPRET/Ei mice exhibit an extreme and dominant resistance to high doses of TNF. In this report, we investigate the response of heterozygous (C57BL/6xSPRET/Ei)F1 mice in different models of inflammatory diseases. Compared with C57BL/6 mice, (B x S)F1 mice are protected against TNF-induced arthritis and are partially protected against allergic asthma in an ovalbumin-induced model. However, these mice display complete susceptibility to TNF-induced inflammatory bowel disease. These results indicate that the SPRET/Ei genome harbors potent dominant antiinflammatory genes that might be relevant for the treatment of certain chronic inflammatory diseases. It is very well possible that different genes are implicated in the different models.


Subject(s)
Inflammation/genetics , Tumor Necrosis Factor-alpha/metabolism , Animals , Arthritis/genetics , Asthma/genetics , Bronchoalveolar Lavage Fluid , Crosses, Genetic , Disease Models, Animal , Female , Genetic Predisposition to Disease , Genome , Heterozygote , Inflammatory Bowel Diseases/genetics , Joints/pathology , Male , Mice , Mice, Inbred C57BL , Ovalbumin/pharmacology
13.
Stem Cells ; 22(4): 441-7, 2004.
Article in English | MEDLINE | ID: mdl-15277691

ABSTRACT

Embryonic stem (ES) cells, which can differentiate into almost all types of cells, have been derived from the house mouse Mus musculus, rat, rabbit, humans, and other species. Transmission of the genotype to the offspring of chimeras has been achieved only with M. musculus ES cells, limiting targeted mutagenesis using ES cells to this species. Mus spretus, which exhibits many genetic polymorphisms with M. musculus, displays dominant resistance to cancer and inflammation, making derived inbred strains very useful in positional cloning and interspecies mapping. We show here for the first time the derivation of ES cells from hybrid blastocysts, obtained by the mating of two different species, namely Mus musculus and Mus spretus, and their use for the generation of chimeric mice that transmit the Mus spretus genotype and phenotype to the offspring. These hybrid ES cells allow the genetic manipulation of Mus spretus, as an alternative to Mus musculus.


Subject(s)
Blastocyst/cytology , Germ Cells/cytology , Stem Cells/cytology , Animals , Chimera , Crosses, Genetic , Humans , Male , Mice , Mice, Inbred C57BL , Muridae , Mutagenesis , Pedigree , Phenotype , Rabbits , Stem Cells/physiology
14.
Proc Natl Acad Sci U S A ; 99(14): 9340-5, 2002 Jul 09.
Article in English | MEDLINE | ID: mdl-12089334

ABSTRACT

Tumor necrosis factor (TNF) is a central mediator in lethal shock and an interesting cytokine for anticancer therapy. To inhibit TNF-induced lethal shock, it is important to identify protective genes. Here we demonstrate that the SPRET/Ei mouse strain, derived from Mus spretus, exhibits an extremely dominant resistance to TNF-induced lethal inflammation. An interspecific backcross experiment revealed that the TNF hyporesponse is linked to loci on chromosomes 2, 6, and 11. Treatment of inoculated tumors with TNF and IFN-gamma leads to regression and a highly reduced toxicity in (C57BL/6 x SPRET/Ei)F(1) mice.


Subject(s)
Shock/etiology , Tumor Necrosis Factor-alpha/toxicity , Animals , Cell Death/drug effects , Drug Resistance , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Genetic Linkage , Interferon-gamma/therapeutic use , Interleukin-6/biosynthesis , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Male , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muridae , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Tumor Necrosis Factor-alpha/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...