Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 10601, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011991

ABSTRACT

Giardia duodenalis is one of the most commonly found intestinal parasites in mammalian hosts. Infections can generally be cleared by mounting an adequate protective immune response that is orchestrated through IL-17A. This study was aimed to investigate if and how the intestinal microbiome affects the protective Th17 response against Giardia by analysing and comparing the immune response following a G. muris and G. duodenalis infection in antibiotic treated and untreated mice. Depletion of the intestinal flora by antibiotic treatment had a severe effect on the infection dynamics of both Giardia species. Not only duration of infection was affected, but also the parasite burden increased significantly. Markers associated with a protective immune response, such as IL-17A and mannose binding lectin 2 were still significantly upregulated following infection in the antibiotic-treated mice, despite the lack of protection. On the other hand, the antibiotic treatment significantly decreased the level of IgA in the intestinal lumen by affecting its transporter and by reducing the number of IgA+ B-cells at the Peyer's patches. Furthermore, the depletion of the gut microbiota by antibiotics also significantly lowered the intestinal motility. The combination of these factors likely results in a decreased clearance of the parasite from the intestinal tract.


Subject(s)
Gastrointestinal Microbiome/immunology , Giardia lamblia/immunology , Immunity , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Bacterial Load , Disease Progression , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Motility/drug effects , Giardia lamblia/drug effects , Giardiasis/drug therapy , Giardiasis/immunology , Giardiasis/microbiology , Giardiasis/parasitology , Immunity/drug effects , Immunoglobulin A/biosynthesis , Interleukin-17/metabolism , Intestines/drug effects , Intestines/immunology , Intestines/microbiology , Intestines/parasitology , Kinetics , Mice, Inbred C57BL , Transcription, Genetic/drug effects
2.
Sci Rep ; 7(1): 13835, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29062028

ABSTRACT

Malaria is a global disease associated with considerable mortality and morbidity. An appropriately balanced immune response is crucial in determining the outcome of malarial infection. The glucocorticoid (GC) metabolising enzyme, 11ß-hydroxysteroid dehydrogenase-1 (11ß-HSD1) converts intrinsically inert GCs into active GCs. 11ß-HSD1 shapes endogenous GC action and is immunomodulatory. We investigated the role of 11ß-HSD1 in two mouse models of malaria. 11ß-HSD1 deficiency did not affect survival after malaria infection, but it increased disease severity and parasitemia in mice infected with Plasmodium chabaudi AS. In contrast, 11ß-HSD1 deficiency rather decreased parasitemia in mice infected with the reticulocyte-restricted parasite Plasmodium berghei NK65 1556Cl1. Malaria-induced antibody production and pathology were unaltered by 11ß-HSD1 deficiency though plasma levels of IL-4, IL-6 and TNF-α were slightly affected by 11ß-HSD1 deficiency, dependent on the infecting parasite. These data suggest that 11ß-HSD1 is not crucial for survival of experimental malaria, but alters its progression in a parasite strain-specific manner.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/deficiency , Malaria/metabolism , Parasitemia/metabolism , Plasmodium chabaudi/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Female , Malaria/genetics , Male , Mice , Mice, Mutant Strains , Parasitemia/genetics , Species Specificity
3.
Sci Rep ; 7(1): 8063, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28808239

ABSTRACT

The glucocorticoid receptor (GR) is a transcription factor of which the underlying gene regulatory mechanisms are complex and incompletely understood. The non-steroidal anti-inflammatory Compound A (CpdA), a selective GR modulating compound in various cell models, has been shown to favour GR-mediated gene repression but not GR-mediated gene activation. Shifting balances towards only a particular subset of GR gene regulatory events may be of benefit in the treatment of inflammatory diseases. We present evidence to support that the combination of CpdA with Dexamethasone (DEX), a classic steroidal GR ligand, can shape GR function towards a unique gene regulatory profile in a cell type-dependent manner. The molecular basis hereof is a changed GR phosphorylation status concomitant with a change in the GR cofactor recruitment profile. We subsequently identified and confirmed the orphan nuclear receptor SHP as a coregulator that is specifically enriched at GR when CpdA and DEX are combined. Combining CpdA with DEX not only leads to stronger suppression of pro-inflammatory gene expression, but also enhanced anti-inflammatory GR target gene expression in epithelial cells, making ligand combination strategies in future a potentially attractive alternative manner of skewing and fine-tuning GR effects towards an improved therapeutic benefit.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Receptors, Glucocorticoid/metabolism , A549 Cells , Animals , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Ligands , Mice , Phosphorylation/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Transcriptional Activation/drug effects
4.
Int J Parasitol ; 45(9-10): 637-46, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25937360

ABSTRACT

Recently we reported the successful vaccination of calves against Cooperia oncophora with a double domain activation-associated secreted protein, purified from the excretory-secretory material of adult stage parasites. In an attempt to elucidate the immune mechanisms involved in protection, the humoral and cell-mediated immune responses following vaccination and infection were compared with non-vaccinated control animals. Antigen-specific IgG1, IgG2 and IgA levels were significantly increased in sera of vaccinated animals post vaccination, whereas no effect was observed for IgM. Antigen-specific intestinal IgG1 levels were significantly increased in the vaccinated animals, whereas no differences were observed for antigen-specific IgA, IgM and IgG2 levels. Upon re-stimulation in vitro with the vaccine antigen, a significant proliferation of both αß- and γδ-T cells, and B cells, collected from mesenteric lymph nodes, was only observed in vaccinated animals. RNA-seq analysis of intestinal tissue yielded a list of 67 genes that were differentially expressed in vaccinated animals following challenge infection, amongst which were several cell adhesion molecules, lectins and glycosyl transferases. A correlation analysis between all immunological and parasitological parameters indicated that intestinal anti-double domain activation-associated secreted protein IgG1 levels correlated negatively with cumulative faecal egg counts and positively with the proportion of L4s and L5s. The proportion of immature stages was also positively correlated with the proliferation of αß T cells. Worm length was negatively correlated with the transcript levels of several lectins and cell adhesion molecules. Overall, the results indicate that intramuscular administration of the vaccine resulted in an immune memory response particularly characterised by increased antigen-specific IgG1 levels in the intestinal mucosa.


Subject(s)
Cattle Diseases/prevention & control , Trichostrongyloidea/immunology , Trichostrongyloidiasis/veterinary , Vaccines/immunology , Animals , Antibodies, Helminth/blood , Antibody Specificity , Antigens, Helminth/immunology , Cattle , Gene Expression Regulation/immunology , Immunoglobulin A/blood , Immunoglobulin G/blood , Injections, Intramuscular , Male , Trichostrongyloidiasis/prevention & control , Vaccination
5.
Mucosal Immunol ; 8(6): 1212-25, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25760421

ABSTRACT

Asthma is a heterogeneous disorder, evidenced by distinct types of inflammation resulting in different responsiveness to therapy with glucocorticoids (GCs). Tumor necrosis factor α (TNFα) is involved in asthma pathogenesis, but anti-TNFα therapies have not proven broadly effective. The effects of anti-TNFα treatment on steroid resistance have never been assessed. We investigated the role of TNFα blockade using etanercept in the responsiveness to GCs in two ovalbumin-based mouse models of airway hyperinflammation. The first model is GC sensitive and T helper type 2 (Th2)/eosinophil driven, whereas the second reflects GC-insensitive, Th1/neutrophil-predominant asthma subphenotypes. We found that TNFα blockade restores the therapeutic effects of GCs in the GC-insensitive model. An adoptive transfer indicated that the TNFα-induced GC insensitivity occurs in the non-myeloid compartment. Early during airway hyperinflammation, mice are GC insensitive specifically at the level of thymic stromal lymphopoietin (Tslp) transcriptional repression, and this insensitivity is reverted when TNFα is neutralized. Interestingly, TSLP knockout mice displayed increased inflammation in the GC-insensitive model, suggesting a limited therapeutic application of TSLP-neutralizing antibodies in subsets of patients suffering from Th2-mediated asthma. In conclusion, we demonstrate that TNFα reduces the responsiveness to GCs in a mouse model of neutrophilic airway inflammation. Thus antagonizing TNFα may offer a new strategy for therapeutic intervention in GC-resistant asthma.


Subject(s)
Asthma/immunology , Drug Resistance/drug effects , Etanercept/pharmacology , Hypersensitivity/immunology , Immunosuppressive Agents/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Anti-Asthmatic Agents/pharmacology , Disease Models, Animal , Female , Glucocorticoids/pharmacology , Immunoassay , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Polymerase Chain Reaction
6.
Ann Rheum Dis ; 69(1): 291-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19204014

ABSTRACT

BACKGROUND: Acquired resistance to glucocorticoids constitutes a major clinical challenge, often overlooked in the search for improved alternatives to classic steroids. We sought to unravel how two glucocorticoid receptor-activating compounds, dexamethasone and Compound A, influence glucocorticoid receptor levels and how this can be correlated to their gene regulatory potential. METHODS: Compound A and dexamethasone were applied in a short-term and long-term treatment protocol. By quantitative PCR analysis in fibroblast-like synoviocytes (FLS) the gene regulatory potential of both compounds in the two experimental conditions was analysed. A parallel Western blot assay revealed the glucocorticoid receptor protein levels in both conditions (ex vivo). In addition, this study examined the effect of systemic administration of dexamethasone and Compound A, in concentrations effective to inhibit collagen-induced arthritis, in DBA/1 mice on glucocorticoid receptor levels (in vivo). RESULTS: Compound A does not induce a homologous downregulation of glucocorticoid receptor in vivo and ex vivo, thereby retaining its anti-inflammatory effects after prolonged treatment in FLS. This is in sharp contrast to dexamethasone, showing a direct link between prolonged dexamethasone treatment, decreasing glucocorticoid receptor levels, and the abolishment of inflammatory gene repression in FLS. It was also observed that the acquired low receptor levels after prolonged dexamethasone treatment are still sufficient to sustain the transactivation of endogenous glucocorticoid-responsive element-driven genes in FLS, a mechanism partly held accountable for the metabolic side-effects. CONCLUSION: Compound A is less likely to evoke therapy resistance, as it does not lead to homologous glucocorticoid receptor downregulation, which is in contrast to classic glucocorticoids.


Subject(s)
Acetates/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Experimental/prevention & control , Arthritis, Rheumatoid/prevention & control , Ethylamines/therapeutic use , Receptors, Glucocorticoid/agonists , Acetates/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Cells, Cultured , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Down-Regulation/drug effects , Drug Evaluation, Preclinical/methods , Drug Resistance , Ethylamines/pharmacology , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Inflammation Mediators/metabolism , Ligands , Male , Mice , Mice, Inbred DBA , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/physiology , Tyramine/analogs & derivatives
7.
Oncogene ; 25(51): 6868-86, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17072333

ABSTRACT

A variety of studies have shown that some activated nuclear receptors (NRs), especially the glucorticoid receptor, the estrogen receptor and peroxisome proliferator-activated receptor, can inhibit the activity of the transcription factor nuclear factor kappaB (NF-kappaB), which plays a key role in the control of genes involved in inflammation, cell proliferation and apoptosis. This review describes the molecular mechanisms of cross-talk between NRs and NF-kappaB and the biological relevance of this cross-talk. The importance and mechanistic aspects of selective NR modulation are discussed. Also included are future research prospects, which will lead to a new era in the field of NR research with the aim of specifically inhibiting NF-kappaB-driven gene expression for anti-inflammatory, anti-tumor and immune-modulatory purposes.


Subject(s)
Hormones/metabolism , NF-kappa B/metabolism , Receptor Cross-Talk , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Humans , Signal Transduction
9.
Biochem Pharmacol ; 60(8): 1185-95, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11007957

ABSTRACT

Interleukin (IL)-6 is a multifunctional cytokine that can be induced by a plethora of chemical or physiological compounds, including the inflammatory cytokines tumor necrosis factor (TNF) and IL-1. The molecule TNF has a trimeric configuration and thus binds to membrane-bound, cellular receptors to initiate cell death mechanisms and signaling pathways leading to gene induction. Previously, we showed that induced clustering of the intracellular domains of the p55 TNF receptor, or of their respective 'death domains' only, is sufficient to activate the nuclear factor kappa B (NF-kappa B) and several mitogen-activated protein kinase (MAPK) pathways. NF-kappa B is the exclusive transcription factor for induction of the IL-6 gene in response to TNF and functions as the final trigger to activate a multiprotein complex, a so-called 'enhanceosome', at the level of the IL-6 promoter. Furthermore, the enhanceosome displays histone acetylation activity, which turned out to be essential for IL-6 gene activation via NF-kappa B. However, activation of NF-kappa B alone is not sufficient for IL-6 gene induction in response to TNF, as inhibition of the coactivated extracellular signal-regulated kinase and p38 MAPK pathways blocks TNF-mediated gene expression. Nevertheless, the transactivating NF-kappa B subunit p65 is not a direct target of MAPK phosphorylation. Thus, we postulated that other components of the enhanceosome complex are sensitive to MAPK cascades and found that MAPK activity is unequivocally linked to the histone acetylation capacity of the enhanceosome to stimulate gene expression in response to TNF. In contrast, glucocorticoid repression of TNF-driven IL-6 gene expression does not depend on abrogation of histone acetyltransferase activity, but originates from interference of the liganded glucocorticoid receptor with the contacts between NF-kappa B p65 and the promoter configuration around the TATA box.


Subject(s)
Gene Expression Regulation , Interleukin-6/genetics , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Enhancer Elements, Genetic/physiology , Humans , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/physiology , Transcriptional Activation
10.
J Neuroimmunol ; 109(1): 16-22, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10969176

ABSTRACT

Glucocorticoids are the most widely used anti-inflammatory and immunomodulatory agents, whose mechanism of action is based mainly on interference with the activity of transcription factors, such as nuclear factor kappaB (NF-kappaB) and activator protein-1 (AP-1). The precise molecular mechanisms of gene repression by glucocorticoids are a controversial matter, due to the existence of many conflicting hypotheses. We discuss the three main paradigms reported in the literature, namely the inhibitor kappaB-alpha (IkappaB-alpha) upregulatory model, the protein-protein interaction model and the competition model.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glucocorticoids/pharmacology , I-kappa B Proteins , Immunosuppressive Agents/pharmacology , Receptors, Glucocorticoid/physiology , Transcription Factors/physiology , Animals , Cyclic AMP Response Element-Binding Protein/physiology , DNA-Binding Proteins/physiology , Humans , NF-KappaB Inhibitor alpha , NF-kappa B/physiology
11.
Proc Natl Acad Sci U S A ; 97(8): 3919-24, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10760263

ABSTRACT

Glucocorticoids (GCs) are used to combat inflammatory diseases. Their beneficial effect relies mainly on the inhibition of NF-kappaB- and/or AP-1-driven proinflammatory gene expression. Previously, we have shown that GCs repress tumor necrosis factor-induced IL-6 gene expression by an NF-kappaB-dependent nuclear mechanism without changing the DNA-binding capacity of NF-kappaB or the expression levels of the cytoplasmic inhibitor of NF-kappaB (IkappaB-alpha). In the present work, we investigate the effect of GC repression on different natural and/or recombinant NF-kappaB-driven reporter gene constructs in the presence of increasing amounts of various coactivator molecules, such as CREB-binding protein (CBP), p300, and SRC-1. We found that GCs maintain their repressive capacities, irrespective of the amount of cofactor present in the cell. Similar results were obtained for the reciprocal transrepression of a GC receptor (GR) element-driven reporter gene by p65. We demonstrate that neither the expression levels of p65 and CBP nor their physical association are affected by activated GR. Using Gal4 chimeras, we show that repression by GCs is specific for p65-mediated transactivation, ruling out competition for limiting nuclear factors as the major underlying mechanism of gene repression. In addition, the transactivation potential of a point-mutated Gal4-p65 variant with a decreased CBP interaction capability is still repressed by GR. Finally, we present evidence that the specificity of GC repression on p65-driven gene expression is codetermined by the TATA box context.


Subject(s)
Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , NF-kappa B/physiology , Saccharomyces cerevisiae Proteins , Transcription, Genetic/drug effects , Base Sequence , DNA Primers , DNA-Binding Proteins , Fungal Proteins/genetics , Gene Expression Regulation/physiology , Mutagenesis, Site-Directed , NF-kappa B/genetics , Promoter Regions, Genetic , TATA Box , Transcription Factor RelA , Transcription Factors/genetics , Transcription, Genetic/physiology , Tumor Necrosis Factor-alpha/physiology
12.
J Biol Chem ; 274(45): 32048-54, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542237

ABSTRACT

Interleukin-6 (IL-6) is a pleiotropic cytokine, whose plasma levels are elevated in inflammatory diseases such as atherosclerosis. We have previously reported that peroxisome proliferator-activated receptor alpha (PPARalpha) ligands (fibrates) lower elevated plasma concentrations of IL-6 in patients with atherosclerosis and inhibit IL-1-stimulated IL-6 secretion by human aortic smooth muscle cells (SMC). Here, we show that aortic explants isolated from PPARalpha-null mice display an exacerbated response to inflammatory stimuli, such as lipopolysaccharide (LPS), as demonstrated by increased IL-6 secretion. Furthermore, fibrate treatment represses IL-6 mRNA levels in LPS-stimulated aortas of PPARalpha wild-type, but not of PPARalpha-null mice, demonstrating a role for PPARalpha in this fibrate action. In human aortic SMC, fibrates inhibit IL-1-induced IL-6 gene expression. Furthermore, activation of PPARalpha represses both c-Jun- and p65-induced transcription of the human IL-6 promoter. Transcriptional interference between PPARalpha and both c-Jun and p65 occurs reciprocally, since c-Jun and p65 also inhibit PPARalpha-mediated activation of a PPAR response element-driven promoter. This transcriptional interference occurs independent of the promoter context as demonstrated by cotransfection experiments using PPARalpha, p65, and c-Jun Gal4 chimeras. Overexpression of the transcriptional coactivator cAMP-responsive element-binding protein-binding protein (CBP) does not relieve PPARalpha-mediated transcriptional repression of p65 and c-Jun. Finally, glutathione S-transferase pull-down experiments demonstrate that PPARalpha physically interacts with c-Jun, p65, and CBP. Altogether these data indicate that fibrates inhibit the vascular inflammatory response via PPARalpha by interfering with the NF-kappaB and AP-1 transactivation capacity involving direct protein-protein interaction with p65 and c-Jun.


Subject(s)
NF-kappa B/physiology , Phlebitis/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factor AP-1/physiology , Transcription Factors/physiology , Animals , COS Cells , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation , Humans , Interleukin-1/pharmacology , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Male , Mice , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Transcription, Genetic , Transfection
13.
J Biol Chem ; 274(45): 32091-8, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542243

ABSTRACT

Expression of the pleiotropic cytokine interleukin (IL)-6 can be stimulated by the proinflammatory cytokine tumor necrosis factor (TNF) and the microbial alkaloid staurosporine (STS). In this report, the transcriptional mechanisms were thoroughly investigated. Whereas transcription factors binding to the activator protein-1-, cAMP-responsive element-, and CAAT enhancer-binding protein-responsive sequences are necessary for gene activation by STS, nuclear factor (NF)-kappaB alone is responsible and sufficient for inducibility by TNF, which reveals distinct signaling pathways for both compounds. At the cofactor level, cAMP-responsive element-binding protein-binding protein (CBP) or p300 potentiate basal and induced IL-6 promoter activation via multiple protein-protein interactions with all transcription factors bound to the promoter DNA. However, the strongest promoter activation relies on the p65 NF-kappaB subunit, which specifically engages CBP/p300 for maximal transcriptional stimulation by its histone acetyltransferase activity. Moreover, treatment of chromatin-integrated promoter constructions with the histone deacetylase inhibitor trichostatin A exclusively potentiates TNF-dependent (i.e. NF-kappaB-mediated) gene activation, while basal or STS-stimulated IL-6 promoter activity remains completely unchanged. Similar observations were recorded with other natural NF-kappaB-driven promoters, namely IL-8 and endothelial leukocyte adhesion molecule (ELAM). We conclude that, within an "enhanceosome-like" structure, NF-kappaB is the central mediator of TNF-induced IL-6 gene expression, involving CBP/p300 and requiring histone acetyltransferase activity.


Subject(s)
Acetyltransferases/metabolism , Interleukin-6/genetics , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Transcriptional Activation , Animals , Cells, Cultured , E1A-Associated p300 Protein , Histone Acetyltransferases , Humans , Hydroxamic Acids/pharmacology , Mice , Staurosporine/pharmacology , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/pharmacology
14.
Mol Pharmacol ; 56(4): 797-806, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10496964

ABSTRACT

Synthetic glucocorticoids (GCs) remain among the most effective agents for the management of chronic inflammatory diseases. However, major side effects severely limit their therapeutic use. Physiologic and therapeutic activities of GCs are mediated by a nuclear receptor belonging to a superfamily of ligand-inducible transcription factors that, in addition to directly regulating their cognate gene programs, can also mutually interfere with other signaling pathways. We recently identified selective ligands of the glucocorticoid receptor that dissociate transactivation from activator protein 1 transrepression, and most importantly retain in vivo anti-inflammatory activity. To further document the mechanisms of action sustaining the observed in vivo activity, we report here on the interference of dissociated GCs with nuclear factor kappaB (NF-kappaB)-driven gene activation. We show that dissociated GCs repress tumor necrosis factor-induced interleukin-6 gene expression by an NF-kappaB-dependent mechanism, without changing the expression level of inhibitor kappaB. The DNA-binding activity of induced NF-kappaB also remained unchanged after stimulation of cells with the various compounds. Evidence for a direct nuclear mechanism of action was obtained by analysis of cell lines constitutively expressing a fusion protein between the DNA-binding domain of the yeast Gal4 protein and the transactivating p65 subunit of NF-kappaB, which was able to efficiently repress a Gal4-dependent luciferase reporter gene upon addition of the dissociated compounds. We therefore conclude that, in addition to dissociating transactivation from activator protein 1 transrepression, dissociated GCs mediate inhibition of NF-kappaB signaling by a mechanism that is independent of inhibitor kappaB induction.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Hydroxycorticosteroids , I-kappa B Proteins , Interleukin-6/genetics , NF-kappa B/physiology , Animals , DNA/metabolism , DNA-Binding Proteins/biosynthesis , Genes, Reporter , HeLa Cells , Hormone Antagonists/pharmacology , Humans , Interleukin-6/metabolism , Mice , Mifepristone/pharmacology , NF-KappaB Inhibitor alpha , Promoter Regions, Genetic , Trans-Activators/physiology , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation , Tumor Cells, Cultured
16.
J Biol Chem ; 273(6): 3285-90, 1998 Feb 06.
Article in English | MEDLINE | ID: mdl-9452444

ABSTRACT

Interleukin-6 (IL-6) is a pleiotropic cytokine, which is involved in inflammatory and immune responses, acute phase reactions, and hematopoiesis. In the mouse fibrosarcoma cell line L929, the nuclear factor (NF)-kappaB plays a crucial role in IL-6 gene expression mediated by tumor necrosis factor (TNF). The levels of the activated factor do not, however, correlate with the variations of IL-6 gene transcription; therefore, other factors and/or regulatory mechanisms presumably modulate the levels of IL-6 mRNA production. Upon analysis of various deletion and point-mutated variants of the human IL-6 gene promoter coupled to a reporter gene, we screened for possible cooperating transcription factors. Even the smallest deletion variant, containing almost exclusively a NF-kappaB-responsive sequence preceding the IL-6 minimal promoter, as well as a recombinant construction containing multiple kappaB-motifs, could still be stimulated with TNF. We observed that the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 was able to repress TNF-stimulated expression of the IL-6 gene, as well as of a kappaB-dependent reporter gene construct, without affecting the levels of NF-kappaB binding to DNA. Furthermore, we clearly show that, using a nuclear Gal4 "one-hybrid" system, the MAPK inhibitors SB203580 and PD0980589 have a direct repressive effect on the transactivation potential of the p65 kappaB subunit. Therefore, we conclude that, in addition to cytoplasmic activation and DNA binding of NF-kappaB, the p38 and extracellular signal-regulated kinase MAPK pathways act as necessary cooperative mechanisms to regulate TNF-induced IL-6 gene expression by modulating the transactivation machinery.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , NF-kappa B/genetics , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Enzyme Activation , Interleukin-6/genetics , Mice , Transcription Factor RelA , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
17.
Proc Natl Acad Sci U S A ; 94(25): 13504-9, 1997 Dec 09.
Article in English | MEDLINE | ID: mdl-9391055

ABSTRACT

Glucocorticoids exert multiple anti-inflammatory activities, one of which is the inhibition of transcription dependent on the nuclear factor (NF)-kappaB. It has been suggested that the effect of dexamethasone (DEX), a glucocorticoid analog, is attributed to an increased production of the inhibitory IkappaB molecule, which in turn would bind and remove activated, DNA-bound NF-kappaB complexes in the cell nucleus. Upon investigating DEX-mediated repression of interleukin-6 expression induced by tumor necrosis factor, DEX treatment was found to act directly on NF-kappaB-dependent transcription, without changing the expression level of IkappaB. Neither the mRNA of IkappaB nor the protein was significantly elevated by a combined treatment with tumor necrosis factor and DEX of murine endothelial or fibroblast cells. The DNA-binding activity of induced NF-kappaB also remained unchanged after stimulation of cells with DEX. Evidence for a direct nuclear mechanism of action was obtained by analysis of cell lines stably expressing a fusion protein between the DNA-binding domain of the yeast Gal4 protein and the transactivating p65 subunit of NF-kappaB. Expression of a Gal4-dependent luciferase reporter gene activated by this nuclear fusion protein was also strongly repressed after addition of DEX. Because the DNA-binding activity of the Gal4 fusion protein was not affected by DEX, it can be concluded that the reduction of gene activation was caused by interference of the activated glucocorticoid receptor with the transactivation potential of the NF-kappaB p65 subunit.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , I-kappa B Proteins , NF-kappa B/metabolism , Transcription, Genetic/drug effects , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Genes, Reporter/drug effects , Interleukin-6/biosynthesis , Interleukin-6/genetics , Mice , Mutagenesis, Site-Directed , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Oligodeoxyribonucleotides/genetics , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...