Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Sci Rep ; 6: 29834, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27431477

ABSTRACT

NLRP3 and ASC are able to form a large multimeric complex called inflammasome in response to a number danger signals. The NLRP3 inflammasome is required for the activation of caspase-1 and subsequent maturation of pro-IL-1ß into active IL-1ß. Although the mechanisms regulating the formation and activity of NLRP3 inflammasome are yet not fully elucidated, data suggest that the assembly of NLRP3 inflammasome requires microtubules to induce the proximity of ASC and NLRP3. In this study we show that microfilaments (F-actin) inhibit NLRP3 inflammasome activity and interact with NLRP3 and ASC. We demonstrate that the inhibition depends on the actin polymerization state but not on the active polymerization process. In ATP- or nigericin-activated macrophages, our data further indicate that Flightless-I (FliI) and leucine-rich repeat FliI-interaction protein 2 (LRRFIP2) are required for the co-localization of NLRP3, ASC and F-actin. We also established that the ability of Ca(2+) to accentuate the activity of NLRP3 inflammasome is abrogated in FliI- and LRRFIP2-knockdown macrophages, suggesting that Ca(2+) signaling requires the presence of FliI and LRRFIP2. Accordingly, we observed that Ca(2+)/FliI-dependent severing of F-actin suppresses F-actin/FliI/LRRFIP2-dependent NLRP3 inflammasome inhibition leading to increase IL-1ß production. Altogether, our results unveil a new function of F-actin in the regulation of NLRP3 inflammasome activity strengthening the importance of cytoskeleton in the regulation of inflammation.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Inflammasomes/metabolism , Microfilament Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Adaptor Proteins, Signal Transducing , Adenosine Triphosphate/pharmacology , Carrier Proteins/genetics , Cytoskeleton/drug effects , Humans , Macrophages/drug effects , Macrophages/metabolism , Microfilament Proteins/genetics , Microscopy, Confocal , Nigericin/pharmacology , Polymerization/drug effects , RNA Interference , Receptors, Cytoplasmic and Nuclear/genetics , THP-1 Cells , Trans-Activators
2.
Ann Rheum Dis ; 74(3): 587-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24326007

ABSTRACT

OBJECTIVES: To investigate the effects and mechanisms of action of high-density lipoproteins (HDL) in monosodium urate (MSU) crystal-induced inflammation -that is, gouty inflammation, in vivo. METHODS: Air pouches raised on the backs of mice were injected with MSU crystals or tumour necrosis factor (TNF) in the presence or absence of HDL and/or interleukin (IL)-1 receptor antagonist (IL-1Ra) for 3 h. Leucocyte count and neutrophil percentage in pouch fluids were measured using a haemocytometer and May-Grünwald-Giemsa staining. The cytokine production and expression in the pouch were measured by ELISA and quantitative RT-PCR. RESULTS: MSU crystals induced leucocyte infiltration, mostly neutrophils, and the release of IL-1ß, IL-6, chemokine (C-X-C motif) ligand 1 (CXCL1), chemokine (C-C motif) ligand 2 (CCL2) and IL-1Ra in pouch fluids. TNF remained under the detection limit. MSU crystals triggered IL-1ß, IL-6 and CXCL1 expression in both pouch exudates and membranes, whereas CCL2 and TNF mRNA were not modulated. The co-injection of MSU crystals and HDL inhibited leucocyte influx by 59% and neutrophil infiltration by 83% and, in turn, both protein and mRNA levels of all assessed proinflammatory cytokines were reduced, but not those of IL-1Ra. Similar results were obtained when mice were injected with MSU crystals pretreated with HDL or TNF instead of crystals. When HDL and IL-1Ra were added together they displayed additional inhibition, suggesting different mechanisms of action. CONCLUSIONS: This study demonstrated that HDL may represent an important factor in the modulation of gouty inflammation by acting on both tissue and infiltrating cells -that is, synovial tissue and synovial fluid cells. HDL display anti-inflammatory activity, in part, by interacting with crystals but also by directly acting on cells.


Subject(s)
Gout , Inflammation/immunology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Lipoproteins, HDL/pharmacology , Subcutaneous Tissue/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Uric Acid/pharmacology , Animals , Back , Chemokine CCL2/drug effects , Chemokine CCL2/immunology , Chemokine CXCL1/drug effects , Chemokine CXCL1/immunology , Disease Models, Animal , Interleukin 1 Receptor Antagonist Protein/drug effects , Interleukin-1beta/drug effects , Interleukin-1beta/immunology , Interleukin-6/immunology , Leukocytes/drug effects , Leukocytes/immunology , Mice , Subcutaneous Tissue/immunology
3.
Behav Brain Res ; 236(1): 131-138, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22974551

ABSTRACT

Studies across and within species suggest that hippocampus size is sexually dimorphic in polygamous species, but not in monogamous species. Although hippocampal volume varies with sex, season and mating system, few studies have simultaneously tested for sex and seasonal differences. Here, we test for sex and seasonal differences in the hippocampal volume of wild Richardson's ground squirrels (Urocitellus richardsonii), a polygamous species that lives in matrilineal, kin-based social groups and has profound sex differences in behavior. Based on the behavior and ecology of this species, we predicted that males would have a significantly larger hippocampus than females and that the hippocampus would be largest in males during the breeding season. Analyses of both absolute and relative volumes of the hippocampus yielded a significant difference between the sexes and seasons as well as an interaction between the two such that non-breeding males have significantly larger hippocampal volumes than breeding males or females from either season. Dentate gyrus, CA1 and CA3 subfield volumes were generally larger in the non-breeding season and in males, but no significant interaction effects were detected. This sex and seasonal variation in hippocampal volume is likely the result of their social organization and male-only food caching behavior during the non-breeding season. The demonstration of a sex and seasonal variation in hippocampal volume suggests that Richardson's ground squirrel may be a useful model for understanding hippocampal plasticity within a natural context.


Subject(s)
Hippocampus/physiology , Sciuridae/physiology , Seasons , Animals , Behavior, Animal/physiology , Body Weight/physiology , Brain Chemistry/physiology , CA1 Region, Hippocampal/anatomy & histology , CA1 Region, Hippocampal/growth & development , CA3 Region, Hippocampal/anatomy & histology , CA3 Region, Hippocampal/growth & development , Dentate Gyrus/anatomy & histology , Dentate Gyrus/growth & development , Estrous Cycle/physiology , Female , Hippocampus/anatomy & histology , Homing Behavior , Linear Models , Male , Sex Characteristics , Sexual Behavior, Animal/physiology , Tissue Fixation
4.
PLoS One ; 6(10): e26293, 2011.
Article in English | MEDLINE | ID: mdl-22022590

ABSTRACT

HYPOTHESIS: T cells modulate the antiviral and inflammatory responses of airway epithelial cells to human rhinoviruses (HRV). METHODS: Differentiated primary human nasal epithelial cells (HNEC) grown on collagen-coated filters were exposed apically to HRV14 for 6 h, washed thoroughly and co-cultured with anti-CD3/CD28 activated T cells added in the basolateral compartment for 40 h. RESULTS: HRV14 did not induce IFNγ, NOS2, CXCL8 and IL-6 in HNEC, but enhanced expression of the T cell attractant CXCL10. On the other hand, HNEC co-cultured with activated T cells produced CXCL10 at a level several orders of magnitude higher than that induced by HRV14. Albeit to a much lower degree, activated T cells also induced CXCL8, IL-6 and NOS2. Anti-IFNγ antibodies and TNF soluble receptor completely blocked CXCL10 upregulation. Furthermore, a significant correlation was observed between epithelial CXCL10 mRNA expression and the amounts of IFNγ and TNF secreted by T cells. Likewise, increasing numbers of T cells to a constant number of HNEC in co-cultures resulted in increasing epithelial CXCL10 production, attaining a plateau at high IFNγ and TNF levels. Hence, HNEC activation by T cells is induced mainly by IFNγ and/or TNF. Activated T cells also markedly inhibited viral replication in HNEC, partially through activation of the nitric oxide pathway. CONCLUSION: Cross-talk between T cells and HNEC results in activation of the latter and increases their contribution to airway inflammation and virus clearance.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/virology , Nose/pathology , Rhinovirus/immunology , T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Clone Cells , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Inflammation , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Rhinovirus/drug effects , Rhinovirus/physiology , Solubility/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Virus Replication/drug effects
5.
CNS Drugs ; 25(5): 401-14, 2011 May.
Article in English | MEDLINE | ID: mdl-21476611

ABSTRACT

Glatiramer acetate is a synthetic, random copolymer widely used as a first-line agent for the treatment of relapsing-remitting multiple sclerosis (MS). While earlier studies primarily attributed its clinical effect to a shift in the cytokine secretion of CD4+ T helper (T(h)) cells, growing evidence in MS and its animal model, experimental autoimmune encephalomyelitis (EAE), suggests that glatiramer acetate treatment is associated with a broader immunomodulatory effect on cells of both the innate and adaptive immune system. To date, glatiramer acetate-mediated modulation of antigen-presenting cells (APC) such as monocytes and dendritic cells, CD4+ T(h) cells, CD8+ T cells, Foxp3+ regulatory T cells and antibody production by plasma cells have been reported; in addition, most recent investigations indicate that glatiramer acetate treatment may also promote regulatory B-cell properties. Experimental evidence suggests that, among these diverse effects, a fostering interplay between anti-inflammatory T-cell populations and regulatory type II APC may be the central axis in glatiramer acetate-mediated immune modulation of CNS autoimmune disease. Besides altering inflammatory processes, glatiramer acetate could exert direct neuroprotective and/or neuroregenerative properties, which could be of relevance for the treatment of MS, but even more so for primarily neurodegenerative disorders, such as Alzheimer's or Parkinson's disease. In this review, we provide a comprehensive and critical overview of established and recent findings aiming to elucidate the complex mechanism of action of glatiramer acetate.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunosuppressive Agents/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis/drug therapy , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Glatiramer Acetate , Humans , Male , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology
6.
Commun Integr Biol ; 4(1): 112-4, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21509198

ABSTRACT

Imbalance in cytokine homeostasis plays an important part in the pathogenesis of various chronic inflammatory diseases. In multiple sclerosis (MS), the pro-inflammatory cytokine interleukin-1ß (IL-1ß) is present in the central nervous system, being expressed mainly in infiltrating macrophages and microglial cells. IL-1ß activity is inhibited by the secreted form of IL-1 receptor antagonist (sIL-1Ra) whose production is increased in patients' blood and induced in human monocytes by IFNß and glatiramer acetate (GA)-both immunomodulators displaying similar therapeutic efficacy in MS. Because intracellular pathways are currently considered as potential therapeutic targets, identification of specific kinases used by both immunomodulators might lead to more specific therapeutic targeting. We addressed the question of intracellular pathways used by IFNß and GA to induce sIL-1Ra in human monocytes in two recent studies. This addendum to these studies aims at discussing common pathways and different elements used by IFNß and GA to induce sIL-1Ra in human monocytes. This pinpoints PI3Kδ activation as a requirement to induce sIL-1Ra production downstream monocyte stimulation by either IFNß or GA. However, the immunomodulators differentially use MEK/ERK pathway to induce sIL-1Ra production in human monocytes. Together, our current studies suggest that PI3Kδ and MEK2 might represent new targets in MS therapy.

7.
Blood ; 117(20): 5523-31, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21330474

ABSTRACT

The presence of antiphospholipid antibodies (aPLAs) is associated with arterial or venous thrombosis and/or recurrent fetal loss. The proposed pathogenic mechanisms for aPLA effects include the inflammatory activation of monocytes and endothelial cells. Toll-like receptors (TLRs) are candidate signaling intermediates. The aim of this study was to investigate the relative contribution of TLR2 and TLR4 in cell activation by aPLAs. Of 32 patient-derived aPLAs, 19 induced an inflammatory activation of human monocytes and umbilical vein endothelial cells (HUVECs). In HUVECs, inflammatory responses to these aPLAs were increased by TNF pretreatment, which increases the expression of TLR2 but not TLR4. Anti-TLR2 but not anti-TLR4 antibodies reduced the aPLA-induced activation of monocytes and HUVECs. aPLAs activated TLR2-expressing human embryonic kidney 293 (HEK293) cells but not TLR4-expressing cells. Binding studies demonstrated an interaction between aPLAs and TLR2 but not TLR4. A role for CD14, a coreceptor for TLR2 and TLR4, can be inferred by observations that anti-CD14 antibodies reduced responses to aPLAs in monocytes, and that responses in HEK293 cells expressing TLR2 and CD14 were greater than in HEK293 cells expressing TLR2 alone. Our results demonstrate a role for TLR2 and CD14 in human endothelial cell and monocyte activation by aPLAs.


Subject(s)
Antibodies, Antiphospholipid/metabolism , Endothelial Cells/immunology , Monocytes/immunology , Toll-Like Receptor 2/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Genes, Reporter , HEK293 Cells , Humans , Lipopolysaccharide Receptors/metabolism , Monocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
8.
J Leukoc Biol ; 88(6): 1191-200, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20837746

ABSTRACT

IFN-ß and sIL-1Ra play crucial roles in the regulation of innate immunity and inflammation. IFN-ß, which is widely used to improve the course of relapsing, remitting multiple sclerosis, induces the production of sIL-1Ra in human monocytes through mechanisms that remain largely unknown. In this study, we identified PI3Kδ and MEK2 as key elements that control sIL-1Ra production in isolated human monocytes activated by IFN-ß. Blockade of MEK2, but not of MEK1, by inhibitors and siRNA prevented IFN-ß-induced PI3Kδ recruitment to the membrane, Akt phosphorylation, and sIL-1Ra production, suggesting that MEK2 acted upstream of PI3Kδ. Furthermore, ERK1/2, the only identified substrates of MEK1/2 to date, are dispensable for sIL-1Ra production in response to IFN-ß stimulation. Upon IFN-ß activation, MEK2 and PI3Kδ are translocated to monocyte membranes. These data suggest that MEK1 and MEK2 display different, nonredundant functions in IFN-ß signaling. That neither MEK1 nor ERK1/2 play a part in this mechanism is also an unexpected finding that gives rise to a better understanding of the MAPK signaling network. Together, these findings demonstrate that IFN-ß triggers an atypical MEK2/PI3Kδ signaling cascade to regulate sIL-1Ra expression in monocytes. The premise that MEK1 and MEK2 play a part in the induction of the proinflammatory cytokine, IL-1ß in human monocytes provides a rationale for an alternative, IFN-ß-mediated pathway to induce/enhance sIL-1Ra production and thus, to dampen inflammation.


Subject(s)
Interferon-beta/pharmacology , Interleukin 1 Receptor Antagonist Protein/biosynthesis , MAP Kinase Kinase 2/physiology , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology , Class I Phosphatidylinositol 3-Kinases , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , MAP Kinase Kinase 1/physiology , Monocytes/drug effects , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
9.
Proc Natl Acad Sci U S A ; 107(41): 17692-7, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20876102

ABSTRACT

Glatiramer acetate (GA), an immunomodulator used in multiple sclerosis (MS) therapy, induces the production of secreted IL-1 receptor antagonist (sIL-1Ra), a natural inhibitor of IL-1ß, in human monocytes, and in turn enhances sIL-1Ra circulating levels in MS patients. GA is a mixture of peptides with random Glu, Lys, Ala, and Tyr sequences of high polarity and hydrophilic nature that is unlikely to cross the blood-brain barrier. In contrast, sIL-1Ra crosses the blood-brain barrier and, in turn, may mediate GA anti-inflammatory activities within the CNS by counteracting IL-1ß activities. Here we identify intracellular signaling pathways induced by GA that control sIL-1Ra expression in human monocytes. By using kinase knockdown and specific inhibitors, we demonstrate that GA induces sIL-1Ra production via the activation of PI3Kδ, Akt, MEK1/2, and ERK1/2, demonstrating that both PI3Kδ/Akt and MEK/ERK pathways rule sIL-1Ra expression in human monocytes. The pathways act in parallel upstream glycogen synthase kinase-3α/ß (GSK3α/ß), the knockdown of which enhances sIL-1Ra production. Together, our findings demonstrate the existence of signal transduction triggered by GA, further highlighting the mechanisms of action of this drug in MS.


Subject(s)
Interleukin 1 Receptor Antagonist Protein/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Peptides/pharmacology , Signal Transduction/drug effects , Blotting, Western , Gene Knockdown Techniques , Glatiramer Acetate , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Interleukin 1 Receptor Antagonist Protein/immunology , Mitogen-Activated Protein Kinase Kinases/metabolism , Monocytes/immunology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/immunology
10.
PLoS One ; 5(7): e11869, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20686620

ABSTRACT

BACKGROUND: Direct cellular contact with stimulated T cells is a potent mechanism that induces cytokine production in human monocytes in the absence of an infectious agent. This mechanism is likely to be relevant to T cell-mediated inflammatory diseases such as rheumatoid arthritis and multiple sclerosis. Microparticles (MP) generated by stimulated T cells (MPT) display similar monocyte activating ability to whole T cells, isolated T cell membranes, or solubilized T cell membranes. We previously demonstrated that high-density lipoproteins (HDL) inhibited T cell contact- and MPT-induced production of IL-1beta but not of its natural inhibitor, the secreted form of IL-1 receptor antagonist (sIL-1Ra). METHODOLOGY/PRINCIPAL FINDINGS: Labeled MPT were used to assess their interaction with monocytes and T lymphocytes by flow cytometry. Similarly, interactions of labeled HDL with monocytes and MPT were assessed by flow cytometry. In parallel, the MPT-induction of IL-1beta and sIL-1Ra production in human monocytes and the effect of HDL were assessed in cell cultures. The results show that MPT, but not MP generated by activated endothelial cells, bond monocytes to trigger cytokine production. MPT did not bind T cells. The inhibition of IL-1beta production by HDL correlated with the inhibition of MPT binding to monocytes. HDL interacted with MPT rather than with monocytes suggesting that they bound the activating factor(s) of T cell surface. Furthermore, prototypical pro-inflammatory cytokines and chemokines such as TNF, IL-6, IL-8, CCL3 and CCL4 displayed a pattern of production induced by MPT and inhibition by HDL similar to IL-1beta, whereas the production of CCL2, like that of sIL-1Ra, was not inhibited by HDL. CONCLUSIONS/SIGNIFICANCE: HDL inhibit both MPT binding to monocytes and the MPT-induced production of some but not all cytokines, shedding new light on the mechanism by which HDL display their anti-inflammatory functions.


Subject(s)
Cell-Derived Microparticles/metabolism , Cytokines/metabolism , Lipoproteins, HDL/pharmacology , Monocytes/drug effects , Monocytes/metabolism , T-Lymphocytes/metabolism , Cell Line , Cell-Derived Microparticles/ultrastructure , Cells, Cultured , Humans , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/metabolism , Microscopy, Electron, Scanning , Monocytes/ultrastructure , T-Lymphocytes/ultrastructure
11.
PLoS One ; 5(2): e9418, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20195532

ABSTRACT

BACKGROUND: Cellular contact with stimulated T cells is a potent inducer of cytokine production in human monocytes and is likely to play a substantial part in chronic/sterile inflammatory diseases. High-density lipoproteins (HDL) specifically inhibit the production of pro-inflammatory cytokines induced by T cell contact. METHODOLOGY/PRINCIPAL FINDINGS: To further elucidate the pro-inflammatory functions of cellular contact with stimulated T cells and its inhibition by HDL, we carried out multiplex and microarray analyses. Multiplex analysis of monocyte supernatant revealed that 12 out of 27 cytokines were induced upon contact with stimulated T cells, which cytokines included IL-1Ra, G-CSF, GM-CSF, IFNgamma, CCL2, CCL5, TNF, IL-1beta, IL-6, IL-8, CCL3, and CCL4, but only the latter six were inhibited by HDL. Microarray analysis showed that 437 out of 54,675 probe sets were enhanced in monocytes activated by contact with stimulated T cells, 164 probe sets (i.e., 38%) being inhibited by HDL. These results were validated by qPCR. Interestingly, the cytokines induced by T cell contact in monocytes comprised IL-1beta, IL-6 but not IL-12, suggesting that this mechanism might favor Th17 polarization, which emphasizes the relevance of this mechanism to chronic inflammatory diseases and highlights the contrast with acute inflammatory conditions that usually involve lipopolysaccharides (LPS). In addition, the expression of miR-155 and production of prostaglandin E(2)-both involved in inflammatory response-were triggered by T cell contact and inhibited in the presence of HDL. CONCLUSIONS/SIGNIFICANCE: These results leave no doubt as to the pro-inflammatory nature of T cell contact-activation of human monocytes and the anti-inflammatory functions of HDL.


Subject(s)
Cytokines/metabolism , Lipoproteins, HDL/pharmacology , Monocytes/metabolism , T-Lymphocytes/drug effects , Cell Line, Tumor , Cells, Cultured , Cytokines/genetics , Dinoprostone/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Humans , Inflammation/genetics , Inflammation/immunology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lymphocyte Activation/immunology , MicroRNAs/genetics , Monocytes/cytology , Monocytes/immunology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Arthritis Res Ther ; 12(1): R23, 2010.
Article in English | MEDLINE | ID: mdl-20149224

ABSTRACT

INTRODUCTION: To investigate whether monosodium urate (MSU) crystals induce the production of CCL2 (monocyte chemoattractant protein-1; MCP-1) in human fibroblast-like synoviocytes (FLS) and whether this mechanism would be affected by high-density lipoproteins (HDL). METHODS: Human FLS isolated from synovial tissue explants were stimulated with MSU crystals (0.01 to 0.5 mg/ml) or interleukin (IL)-1beta (10 pg/ml) in the presence or absence of HDL (50 and 100 microg/ml). The production and expression of CCL2 was evaluated with ELISA, confocal microscopy, immunofluorescence microscopy, chemotaxis assay, and real-time quantitative PCR. RESULTS: Exposure of FLS to MSU crystals induced CCL2 accumulation in culture medium in a dose- and time-dependent manner, reaching a plateau at 50 to 75 microg/ml MSU crystals and 20 to 24 hours. Although low, the induced CCL2 levels were sufficient to trigger mononuclear cell migration. In resting FLS, CCL2 was localized in small cytoplasmic vesicles whose number diminished with MSU crystal stimulation. Concomitantly, MSU crystals triggered the induction of CCL2 mRNA expression. All these processes were inhibited by HDL, which cause a 50% decrease in CCL2 mRNA levels and a dose-dependent inhibition of the release of CCL2. Similar results were obtained when FLS were pretreated with HDL and washed before activation by MSU crystals or IL-1beta, suggesting a direct effect of HDL on the FLS activation state. CONCLUSIONS: The present results demonstrate that MSU crystals induce FLS to release CCL2 that is stored in vesicles in resting conditions. This mechanism is inhibited by HDL, which may limit the inflammatory process by diminishing CCL2 production and, in turn, monocytes/macrophages recruitment in joints. This study confirms the antiinflammatory functions of HDL, which might play a part in the limitation of acute gout attack.


Subject(s)
Chemokine CCL2/biosynthesis , Fibroblasts/metabolism , Lipoproteins, HDL/metabolism , Synovial Membrane/metabolism , Uric Acid/metabolism , Cells, Cultured , Crystallization , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Fibroblasts/drug effects , Gout/metabolism , Humans , Microscopy, Confocal , Synovial Membrane/cytology , Synovial Membrane/drug effects , Uric Acid/adverse effects
13.
Proc Natl Acad Sci U S A ; 106(11): 4355-9, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19255448

ABSTRACT

Mechanisms of action as well as cellular targets of glatiramer acetate (GA) in multiple sclerosis (MS) are still not entirely understood. IL-1beta is present in CNS-infiltrating macrophages and microglial cells and is an important mediator of inflammation in experimental autoimmune encephalitis (EAE), the MS animal model. A natural inhibitor of IL-1beta, the secreted form of IL-1 receptor antagonist (sIL-1Ra) improves EAE disease course. In this study we examined the effects of GA on the IL-1 system. In vivo, GA treatment enhanced sIL-1Ra blood levels in both EAE mice and patients with MS, whereas IL-1beta levels remained undetectable. In vitro, GA per se induced the transcription and production of sIL-1Ra in isolated human monocytes. Furthermore, in T cell contact-activated monocytes, a mechanism relevant to chronic inflammation, GA strongly diminished the expression of IL-1beta and enhanced that of sIL-1Ra. This contrasts with the effect of GA in monocytes activated upon acute inflammatory conditions. Indeed, in LPS-activated monocytes, IL-1beta and sIL-1Ra production were increased in the presence of GA. These results demonstrate that, in chronic inflammatory conditions, GA enhances circulating sIL-1Ra levels and directly affects monocytes by triggering a bias toward a less inflammatory profile, increasing sIL-1Ra while diminishing IL-1beta production. This study sheds light on a mechanism that is likely to participate in the therapeutic effects of GA in MS.


Subject(s)
Interleukin 1 Receptor Antagonist Protein/drug effects , Interleukin-1beta/drug effects , Monocytes/immunology , Multiple Sclerosis/drug therapy , Peptides/pharmacology , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Glatiramer Acetate , Humans , Immunosuppressive Agents/pharmacology , Inflammation/drug therapy , Interleukin 1 Receptor Antagonist Protein/blood , Mice , Multiple Sclerosis/immunology , Transcription, Genetic
14.
Mol Immunol ; 45(12): 3419-27, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18471882

ABSTRACT

Deregulated production of cytokines, including IL-1beta, IL-6 and TNF plays an important role in chronic inflammation. Relevant to this condition, direct cellular contact with stimulated T cells is a potent inducer of cytokine production in human monocytes/macrophages. We previously demonstrated that PI3Ks regulate differential production of IL-1beta and its specific inhibitor secreted IL-1 receptor antagonist (sIL-1Ra) by human monocytes. Here we show that in contrast with PI3Kalpha, beta and gamma, PI3Kdelta accounts for most of the PI3K-dependent signaling ruling the production of IL-1beta, IL-6, TNF and sIL-1Ra in monocytes activated by cellular contact with stimulated T cells (mimicked by CHAPS-solubilized membranes of stimulated T cells, CE sHUT) and lipopolysaccharides (LPS); the latter stimuli being relevant to chronic/sterile and acute/infectious inflammation, respectively. Interestingly, PI3Kdelta activity dampened the production of pro-inflammatory cytokines in LPS-activated monocytes, but induced it in CE sHUT-activated cells. In both CE sHUT- and LPS-activated monocytes PI3Kdelta regulated cytokine transcript expression through the phosphorylation/inactivation of glycogen synthase kinase-3beta (GSK3beta). The blockade of GSK3beta displayed inverse effects to those of PI3Kdelta blockade. Thus, by displaying opposite functions in conditions mimicking chronic/sterile and acute/infectious inflammation, i.e., by repressing pro-inflammatory cytokine expression in LPS-activated monocytes but inducing such mediators in T cell contact-activated monocytes, PI3Kdelta represents a potential therapeutic target specific to chronic/sterile inflammatory conditions.


Subject(s)
Cytokines/biosynthesis , Inflammation/enzymology , Monocytes/enzymology , Monocytes/immunology , Phosphatidylinositol 3-Kinases/metabolism , Acute Disease , Cell Line , Cell Separation , Chronic Disease , Class I Phosphatidylinositol 3-Kinases , Class Ib Phosphatidylinositol 3-Kinase , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Isoenzymes/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Monocytes/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis
15.
J Leukoc Biol ; 83(4): 921-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18223103

ABSTRACT

Imbalance in cytokine homeostasis plays an important part in the pathogenesis of chronic inflammatory diseases such as multiple sclerosis and rheumatoid arthritis. We demonstrated that T cells might exert a pathological effect through direct cellular contact with human monocytes/macrophages, inducing a massive up-regulation of the prototypical proinflammatory cytokines IL-1beta and TNF. This mechanism that might be implicated in chronic inflammation is specifically inhibited by high-density lipoproteins (HDL). Like many other stimuli, besides proinflammatory cytokines, the contact-mediated activation of monocytes induces the production of cytokine inhibitors such as the secreted form of the IL-1 receptor antagonist (sIL-1Ra). The present study demonstrates that stimulated T cells generate microparticles (MP) that induce the production of TNF, IL-1beta, and sIL-1Ra in human monocytes; the production of TNF and IL-1beta but not that of sIL-1Ra is inhibited in the presence of HDL. The results were similar when monocytes were stimulated by whole membranes of T cells or soluble extracts of the latter. This suggests that MP carry similar monocyte-activating factors to cells from which they originate. Thus, by releasing MP, T cells might convey surface molecules similar to those involved in the activation of monocytes by cellular contact. By extension, MP might affect the activity of cells, which are usually not in direct contact with T cells at the inflammatory site. Furthermore, this study demonstrates that HDL exert an anti-inflammatory effect in nonseptic activation of human monocytes, not only by inhibiting the production of IL-1beta and TNF but also, by leaving sIL-1Ra production unchanged.


Subject(s)
Cytokines/biosynthesis , Lipoproteins, HDL/pharmacology , Lymphocyte Activation , Monocytes/immunology , T-Lymphocytes/immunology , Cell Line , Cytokines/blood , Humans , Inflammation/physiopathology , Inflammation/prevention & control , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin-1beta/blood , RNA, Messenger/genetics , Reference Values , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
16.
Methods Mol Med ; 136: 139-48, 2007.
Article in English | MEDLINE | ID: mdl-17983145

ABSTRACT

The production of cytokines and other inflammatory products in chronic/sterile inflammatory disorders such as rheumatoid arthritis might be induced in monocytes/macrophages by direct cellular contact with stimulated T-cells. Studies of cell-cell interactions are usually complicated by the simultaneous presence of at least two viable cell types. To obviate this problem, we developed strategies allowing only interactions between stimulated T-cell surface molecules and the monocytes/macrophages, and any products secreted into the medium or mRNA are necessarily of monocytic origin. This chapter aims at reviewing and describing the latter methods and strategies.


Subject(s)
Cell Separation/methods , Macrophages/immunology , Monocytes/immunology , T-Lymphocytes/immunology , Cytokines/immunology , Humans , Lymphocyte Activation , Macrophages/cytology , Monocytes/cytology , RNA, Messenger/analysis , T-Lymphocytes/cytology
17.
J Immunol ; 178(1): 446-54, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17182583

ABSTRACT

The unbalanced production of IL-1beta and its natural, specific inhibitor, the secreted IL-1R antagonist (sIL-1Ra), plays an important role in chronic/sterile inflammation. Relevant to this condition is direct cellular contact with stimulated T cells which is a potent inducer of cytokine production in human monocytes/macrophages. We previously demonstrated that activation of PI3Ks is a prerequisite of the transcription of the sIL-1Ra gene in human monocytes activated by IFN-beta. In this study, we addressed the question of PI3K involvement in the production of IL-1beta and sIL-1Ra in monocytes activated by cellular contact with stimulated T cells (mimicked by CHAPS-solubilized membranes of stimulated T cells (CE(sHUT))), and a crude preparation of LPS, to compare stimuli relevant to chronic/sterile and acute/infectious inflammation, respectively. In monocytes activated by either CE(sHUT) or LPS, the inhibition of PI3Ks abrogated sIL-1Ra transcript expression and sIL-1Ra production, demonstrating that PI3Ks control the induction of sIL-1Ra gene transcription. In contrast, PI3K inhibition increased the production of IL-1beta protein in both CE(sHUT)- and LPS-activated monocytes, the enhancement being drastically higher in the former. This was not due to changes in IL-1beta mRNA steady-state levels or transcript stability, but to the involvement of PI3Ks in the repression of IL-1beta secretion. The downstream PI3K effector, Akt, was implicated in this process. The present results demonstrate that PI3Ks are involved in the inhibition of IL-1beta secretion and in the induction of sIL-1Ra production in human blood monocytes by controlling different mechanisms in conditions mimicking chronic/sterile (CE(sHUT)) and acute/infectious (LPS) inflammation.


Subject(s)
Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/metabolism , Monocytes/immunology , Phosphatidylinositol 3-Kinases/metabolism , Cell Line , Down-Regulation , Gene Expression Regulation , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/genetics , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Stability , RNA, Messenger/metabolism , T-Lymphocytes/immunology , Transcription, Genetic , Up-Regulation
18.
Best Pract Res Clin Rheumatol ; 20(5): 879-96, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16980212

ABSTRACT

Inflammation is an important homeostatic mechanism that limits the effects of infectious agents. However, inflammation might be self-damaging and therefore has to be tightly controlled or even abolished by the organism. Interleukin 1 (IL-1) is a crucial mediator of the inflammatory response, playing an important part in the body's natural responses and the development of pathological conditions leading to chronic inflammation. While IL-1 production may be decreased or its effects limited by so-called anti-inflammatory cytokines, in vitro IL-1 inflammatory effects are inhibited and can be abolished by one particularly powerful inhibitor, IL-1 receptor antagonist (IL-1Ra). Recent research has shown that in the processes of rheumatoid arthritis (RA) IL-1 is one of the pivotal cytokines in initiating disease, and IL-1Ra has been shown conclusively to block its effects. In laboratory and animal studies the inhibition of IL-1 by either antibodies to IL-1 or IL-1Ra proved beneficial to the outcome. Because of its beneficial effects in many animal disease models, IL-1Ra has been used as a therapeutic agent in human patients. The recombinant form of IL-1Ra, anakinra (Kineret, Amgen) failed to show beneficial effects in septic shock and displays weak effects in RA patients. However, IL-1 blockade by anakinra is dramatically effective in systemic-onset juvenile idiopathic arthritis, in adult Still's disease and in several autoinflammatory disorders, most of the latter being caused by mutations of proteins controlling IL-1beta secretion. Importantly, to be efficacious, anakinra required daily injections, suggesting that administered IL-1Ra displays very short-term effects. Better IL-1 antagonists are in the process of being developed.


Subject(s)
Arthritis/drug therapy , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/physiopathology , Disease Models, Animal , Humans , Interleukin 1 Receptor Antagonist Protein/physiology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1/antagonists & inhibitors , Interleukin-1/physiology , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Receptors, Interleukin/antagonists & inhibitors , Receptors, Interleukin/drug effects , Receptors, Interleukin/physiology
19.
Expert Opin Biol Ther ; 5 Suppl 1: S83-96, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16187943

ABSTRACT

An imbalance in cytokine homeostasis is considered to play a major part in the pathogenesis of immuno-inflammatory diseases. Since the identification and cloning of cytokines and their receptors, therapeutic approaches have been developed with the purpose of impeding the interaction between the ligand (cytokine) and its specific receptor, or interactions that involve the use of anti-inflammatory cytokines to switch off inflammation. Although some diseases have been treated successfully with cytokines or anticytokines (i.e., anti-TNF, and to a lesser extent recombinant IL-1 receptor antagonist, in rheumatoid arthritis; IFN-beta in multiple sclerosis), the fact remains that these therapies do not abrogate the concomitant use of steroids or immunosuppressive drugs, and that a significant percentage of patients do not respond to such therapies; these are important limitations. The identification of signalling pathways preferentially used in inflammatory conditions has boosted approaches that target these intracellular mechanisms. This review examines the different therapeutic approaches that may be considered for the treatment of immuno-inflammatory diseases, and discusses the advantages and disadvantages of targeting extracellular or intracellular mechanisms.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/antagonists & inhibitors , Immunosuppressive Agents/pharmacology , Receptors, Cytokine/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Clinical Trials as Topic , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Immune System Diseases/drug therapy , Immune System Diseases/metabolism , Immunosuppressive Agents/therapeutic use , Inflammation/metabolism , Interleukin 1 Receptor Antagonist Protein , Receptors, Cytokine/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Sialoglycoproteins/pharmacology , Sialoglycoproteins/therapeutic use , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
J Immunol ; 174(5): 2974-80, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15728510

ABSTRACT

IFN-beta induces the production of secreted IL-1R antagonist (sIL-1Ra) without triggering synthesis of the agonist IL-1beta in human monocytes. This might account for its anti-inflammatory properties. Canonically, IFN-beta signals through activation of JAK/STAT pathway, although PI3K and MAPK have also been involved. In this study, the role of PI3K, MEK1, and STAT1 in IFN-beta-induced sIL-1Ra production is investigated in freshly isolated human blood monocytes. PI3K, but not MEK1 activation is essential for sIL-1Ra production in monocytes treated with IFN-beta, as demonstrated by using the respective inhibitors of PI3K and MEK1, Ly294002 and PD98059. The use of cycloheximide and actinomycin D shows that sIL-1Ra was an immediate early gene induced by IFN-beta and that PI3K was controlling sIL-1Ra gene transcription. Although both inhibitors of PI3K and MEK1 diminished the Ser(727) phosphorylation of STAT1 induced by IFN-beta, only Ly294002 inhibited sIL-1Ra production. Furthermore, the inhibition of STAT1-Ser(727) phosphorylation by Ly294002 did not affect STAT1 translocation, suggesting that STAT1 was not involved in sIL-1Ra gene induction. This was confirmed in monocytes that were transfected with small interfering RNA specifically targeting STAT1. Indeed, monocytes in which effective STAT1 gene knockdown was achieved were fully responsive to IFN-beta in terms of sIL-1Ra production. Taken together, the present data demonstrate that the induction of sIL-1Ra transcription and production by IFN-beta in human monocytes involved PI3K, but not STAT1 activation.


Subject(s)
DNA-Binding Proteins , Interferon-beta/pharmacology , Monocytes/immunology , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Sialoglycoproteins/biosynthesis , Signal Transduction/immunology , Trans-Activators , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , Chromones/pharmacology , Consensus Sequence , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Enzyme Activation/immunology , Humans , Interleukin 1 Receptor Antagonist Protein , MAP Kinase Kinase 1/metabolism , Monocytes/enzymology , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Binding/drug effects , Protein Binding/immunology , STAT1 Transcription Factor , Serine/metabolism , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Signal Transduction/drug effects , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Trans-Activators/physiology , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...