Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Crit Rev Immunol ; 34(4): 315-45, 2014.
Article in English | MEDLINE | ID: mdl-24941159

ABSTRACT

IL-10 is an important anti-inflammatory cytokine that plays important roles in controlling inflammatory responses and keeping the immune system in check following activation. Loss of IL-10 function in mice or humans results in the development of inflammatory bowel disease in response to an elevated immune response to the gut flora. IL-10 also acts to prevent excessive inflammation during the course of infection and has been implicated in a variety of autoimmune conditions. In response to inflammatory signals, IL-10 can be produced by a number of immune cells including T cells, B cells, macrophages, and dendritic cells. Distinct mechanisms control the production of IL-10 in these different cells types. In this review, we describe recent studies that have looked at the signaling pathways that regulate IL-10 production in these cells. Given the number of cell types that produce IL-10, it is perhaps not surprising that the in vivo source of IL-10 can vary in different immune models. We also describe how work using conditional IL-10 knockout mice or adoptive transfer of IL-10-deficient cells has begun to further our understanding regarding which specific immune cells are required for IL-10 production in vivo under different conditions.


Subject(s)
Gene Expression Regulation , Interleukin-10/genetics , Transcription, Genetic , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Interleukin-10/metabolism , Intracellular Space/metabolism , Macrophages/immunology , Macrophages/metabolism , Organ Specificity/genetics , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
FEBS Lett ; 587(10): 1496-503, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23542035

ABSTRACT

Chemokines, including MCP-1, are crucial to mounting an effective immune response due to their ability to recruit other immune cells. We show that sustained LPS or poly(I:C)-stimulated MCP-1 production requires an IFNß-mediated feedback loop. Consistent with this, exogenous IFNß was able to induce MCP-1 transcription in the absence of other stimuli. Blocking IFNß signaling with Ruxolitinib, a JAK inhibitor, inhibited MCP-1 transcription. The MCP-1 promoter contains potential STAT binding sites and we demonstrate that STAT1 is recruited upon IFNß stimulation. Furthermore we find that IL-10 knockout increases MCP-1 production in response to LPS, which may reflect an ability of IL-10 to repress IFNß production. Overall, these results show the importance of the balance between IFNß and IL-10 in the regulation of MCP-1.


Subject(s)
Autocrine Communication/physiology , Chemokine CCL2/genetics , Feedback, Physiological/physiology , Interferon-beta/physiology , Macrophages/metabolism , Toll-Like Receptors/physiology , Animals , Autocrine Communication/drug effects , Autocrine Communication/genetics , Cells, Cultured , Chemokine CCL2/metabolism , Feedback, Physiological/drug effects , Interferon-beta/metabolism , Interferon-beta/pharmacology , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-10/physiology , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Toll-Like Receptors/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
3.
PLoS One ; 8(3): e60086, 2013.
Article in English | MEDLINE | ID: mdl-23533666

ABSTRACT

In response to infection by fungal pathogens, the innate immune system recognises specific fungal pathogen associated molecular patterns (PAMPs) via pattern recognition receptors including the C-type lectin dectin-1 and members of the Toll Like Receptor (TLR) family. Stimulation of these receptors leads to the induction of both pro- and anti-inflammatory cytokines. The protein kinases MSK1 and 2 are known to be important in limiting inflammatory cytokine production by macrophages in response to the TLR4 agonist LPS. In this study we show that MSKs are also activated in macrophages by the fungal derived ligand zymosan, as well as the dectin-1 specific agonists curdlan and depleted zymosan, via the ERK1/2 and p38α MAPK pathways. Furthermore, we show that MSKs regulate dectin-1 induced IL-10 production, and that this regulation is dependent on the ability of MSKs to phosphorylate the transcription factor CREB. IL-10 secreted in response to zymosan was able to promote STAT3 phosphorylation via an autocrine feedback loop. Consistent with the decreased IL-10 secretion in MSK1/2 knockout macrophages, these cells also had decreased STAT3 tyrosine phosphorylation relative to wild type controls after stimulation with zymosan. We further show that the reduction in IL-10 production in the MSK1/2 macrophages results in increased secretion of IL-12p40 in response to zymosan relative to wild type controls. The production of high levels of IL-10 but low levels of IL-12 has previously been associated with an M2b or 'regulatory' macrophage phenotype, which was initially described in macrophages stimulated with a combination of immune complexes and LPS. We found that zymosan, via dectin-1 activation, also leads to the expression of SphK1 and LIGHT, markers of a regulatory like phenotype in mouse macrophages. The expression of these makers was further reinforced by the high level of IL-10 secreted in response to zymosan stimulation.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Interleukin-10/metabolism , Lectins, C-Type/metabolism , Macrophages/drug effects , Macrophages/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Immunoblotting , Lectins, C-Type/genetics , Mice , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction/drug effects , Zymosan/pharmacology
4.
Mol Cell Biol ; 33(7): 1456-67, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23382072

ABSTRACT

Prostaglandin production is catalyzed by cyclooxygenase 2 (cox-2). We demonstrate here that MSK1 and MSK2 (MSK1/2) can exert control on the induction of cox-2 mRNA by Toll-like receptor (TLR) agonists. In the initial phase of cox-2 induction, MSK1/2 knockout macrophages confirmed a role for MSK in the positive regulation of transcription. However, at later time points both lipopolysaccharide (LPS)-induced prostaglandin and cox-2 protein levels were increased in MSK1/2 knockout. Further analysis found that while MSKs promoted cox-2 mRNA transcription, following longer LPS stimulation MSKs also promoted degradation of cox-2 mRNA. This was found to be the result of an interleukin 10 (IL-10) feedback mechanism, with endogenously produced IL-10 promoting cox-2 degradation. The ability of IL-10 to do this was dependent on the mRNA binding protein TTP through a p38/MK2-mediated mechanism. As MSKs regulate IL-10 production in response to LPS, MSK1/2 knockout results in reduced IL-10 secretion and therefore reduced feedback from IL-10 on cox-2 mRNA stability. Following LPS stimulation, this increased mRNA stability correlated to an elevated induction of both of cox-2 protein and prostaglandin secretion in MSK1/2 knockout macrophages relative to that in wild-type cells. This was not restricted to isolated macrophages, as a similar effect of MSK1/2 knockout was seen on plasma prostaglandin E2 (PGE2) levels following intraperitoneal injection of LPS.


Subject(s)
Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Prostaglandins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Interleukin-10/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Prostaglandins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteolysis , RNA Stability , RNA, Messenger/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Immunol ; 190(2): 565-77, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23241891

ABSTRACT

The polarization of macrophages into a regulatory-like phenotype and the production of IL-10 plays an important role in the resolution of inflammation. We show in this study that PGE(2), in combination with LPS, is able to promote an anti-inflammatory phenotype in macrophages characterized by high expression of IL-10 and the regulatory markers SPHK1 and LIGHT via a protein kinase A-dependent pathway. Both TLR agonists and PGE(2) promote the phosphorylation of the transcription factor CREB on Ser(133). However, although CREB regulates IL-10 transcription, the mutation of Ser(133) to Ala in the endogenous CREB gene did not prevent the ability of PGE(2) to promote IL-10 transcription. Instead, we demonstrate that protein kinase A regulates the phosphorylation of salt-inducible kinase 2 on Ser(343), inhibiting its ability to phosphorylate CREB-regulated transcription coactivator 3 in cells. This in turn allows CREB-regulated transcription coactivator 3 to translocate to the nucleus where it serves as a coactivator with the transcription factor CREB to induce IL-10 transcription. In line with this, we find that either genetic or pharmacological inhibition of salt-inducible kinases mimics the effect of PGE(2) on IL-10 production.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/pharmacology , Interleukin-10/biosynthesis , Macrophages/drug effects , Macrophages/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Interleukin-10/genetics , Mice , Phenotype , Phosphorylation/drug effects , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
6.
Proc Natl Acad Sci U S A ; 109(42): 16986-91, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23033494

ABSTRACT

Macrophages acquire strikingly different properties that enable them to play key roles during the initiation, propagation, and resolution of inflammation. Classically activated (M1) macrophages produce proinflammatory mediators to combat invading pathogens and respond to tissue damage in the host, whereas regulatory macrophages (M2b) produce high levels of anti-inflammatory molecules, such as IL-10, and low levels of proinflammatory cytokines, like IL-12, and are important for the resolution of inflammatory responses. A central problem in this area is to understand how the formation of regulatory macrophages can be promoted at sites of inflammation to prevent and/or alleviate chronic inflammatory and autoimmune diseases. Here, we demonstrate that the salt-inducible kinases (SIKs) restrict the formation of regulatory macrophages and that their inhibition induces striking increases in many of the characteristic markers of regulatory macrophages, greatly stimulating the production of IL-10 and other anti-inflammatory molecules. We show that SIK inhibitors elevate IL-10 production by inducing the dephosphorylation of cAMP response element-binding protein (CREB)-regulated transcriptional coactivator (CRTC) 3, its dissociation from 14-3-3 proteins and its translocation to the nucleus where it enhances a gene transcription program controlled by CREB. Importantly, the effects of SIK inhibitors on IL-10 production are lost in macrophages that express a drug-resistant mutant of SIK2. These findings identify SIKs as a key molecular switch whose inhibition reprograms macrophages to an anti-inflammatory phenotype. The remarkable effects of SIK inhibitors on macrophage function suggest that drugs that target these protein kinases may have therapeutic potential for the treatment of inflammatory and autoimmune diseases.


Subject(s)
Cyclobutanes/pharmacology , Indans/pharmacology , Inflammation/immunology , Macrophages/immunology , Morpholines/pharmacology , Phenylurea Compounds/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/pharmacology , Transcription Factors/metabolism , Analysis of Variance , Animals , Cell Line , Cyclobutanes/chemical synthesis , Cytokines/metabolism , DNA Primers/genetics , DNA, Complementary/genetics , Fluorescent Antibody Technique , Immunoblotting , Interleukin-10/genetics , Interleukin-10/metabolism , Macrophages/metabolism , Magnetic Resonance Spectroscopy , Mice , Mice, Knockout , Molecular Structure , Morpholines/chemical synthesis , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/chemistry , Phosphorylation , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Proteomics , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , RNA Interference
7.
J Immunol ; 189(6): 2784-92, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22904308

ABSTRACT

Macrophages are an important source of cytokines following infection. Stimulation of macrophages with TLR agonists results in the secretion of TNF-α, IL-6, and IL-12, and the production of these cytokines is controlled by multiple feedback pathways. Macrophages also produce IL-10, which acts to inhibit proinflammatory cytokine production by macrophages via a JAK/STAT3-dependent pathway. We show in this paper that, Ruxolitinib, a recently described selective inhibitor of JAKs, increases TNF, IL-6, and IL-12 secretion in mouse bone marrow-derived macrophages stimulated with LPS. This effect is largely due to its ability to block IL-10-mediated feedback inhibition on cytokine transcription in macrophages. Similar results were also obtained with a second structurally unrelated Jak inhibitor, Tofacitinib. In addition, LPS induced the production of IFN-ß, which was then able to activate JAKs in macrophages, resulting in the stimulation of STAT1 phosphorylation. The initial induction of IL-10 was independent of JAK signaling; however, inhibition of JAKs did reduce IL-10 secretion at later time points. This reflected a requirement for the IFN-ß feedback loop to sustain IL-10 transcription following LPS stimulation. In addition to IL-10, IFN-ß also helped sustain IL-6 and IL-12 transcription. Overall, these results suggest that inhibition of JAKs may increase the inflammatory potential of macrophages stimulated with TLR4 agonists.


Subject(s)
Bone Marrow Cells/immunology , Cytokines/biosynthesis , Feedback , Interleukin-10/antagonists & inhibitors , Janus Kinases/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/immunology , Up-Regulation/immunology , Animals , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Cells, Cultured , Inflammation Mediators/physiology , Interferon Type I/physiology , Interleukin-10/physiology , Janus Kinases/physiology , Macrophages/enzymology , Macrophages/pathology , Mice , Nitriles , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction/immunology , Toll-Like Receptors/physiology
8.
Proc Natl Acad Sci U S A ; 109(28): 11200-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733747

ABSTRACT

On the basis mainly of pharmacological experiments, the p38α MAP kinase isoform has been established as an important regulator of immune and inflammatory responses. However, the role of the related p38γ and p38δ kinases has remained unclear. Here, we show that deletion of p38γ and p38δ impaired the innate immune response to lipopolysaccharide (LPS), a Toll-like receptor 4 (TLR4) ligand, by blocking the extracellular signal-regulated kinase 1/2 (ERK1/2) activation in macrophages and dendritic cells. p38γ and p38δ were necessary to maintain steady-state levels of tumor progression locus 2 (TPL2), the MKK kinase that mediates ERK1/2 activation after TLR4 stimulation. TNFα, IL-1ß, and IL-10 production were reduced in LPS-stimulated macrophages from p38γ/δ-null mice, whereas IL-12 and IFNß production increased, in accordance with the known effects of TPL2/ERK1/2 signaling on the induction of these cytokines. Furthermore, p38γ/δ-deficient mice were less sensitive than controls to LPS-induced septic shock, showing lower TNFα and IL-1ß levels after challenge. Together, our results establish p38γ and p38δ as key components in innate immune responses.


Subject(s)
Cytokines/metabolism , Gene Expression Regulation , Mitogen-Activated Protein Kinase 13/chemistry , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/chemistry , Animals , Cattle , Cells, Cultured , Culture Media, Conditioned/pharmacology , Gene Deletion , Humans , Immunity, Innate , MAP Kinase Signaling System , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Protein Isoforms , Shock, Septic/metabolism
9.
Biochem J ; 435(3): 755-69, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21323643

ABSTRACT

cAMP-specific PDE (phosphodiesterase) 4 isoforms underpin compartmentalized cAMP signalling in mammalian cells through targeting to specific signalling complexes. Their importance is apparent as PDE4 selective inhibitors exert profound anti-inflammatory effects and act as cognitive enhancers. The p38 MAPK (mitogen-activated protein kinase) signalling cascade is a key signal transduction pathway involved in the control of cellular immune, inflammatory and stress responses. In the present study, we show that PDE4A5 is phosphorylated at Ser147, within the regulatory UCR1 (ultraconserved region 1) domain conserved among PDE4 long isoforms, by MK2 (MAPK-activated protein kinase 2, also called MAPKAPK2). Phosphorylation by MK2, although not altering PDE4A5 activity, markedly attenuates PDE4A5 activation through phosphorylation by protein kinase A. This modification confers the amplification of intracellular cAMP accumulation in response to adenylate cyclase activation by attenuating a major desensitization system to cAMP. Such reprogramming of cAMP accumulation is recapitulated in wild-type primary macrophages, but not MK2/3-null macrophages. Phosphorylation by MK2 also triggers a conformational change in PDE4A5 that attenuates PDE4A5 interaction with proteins whose binding involves UCR2, such as DISC1 (disrupted in schizophrenia 1) and AIP (aryl hydrocarbon receptor-interacting protein), but not the UCR2-independent interacting scaffold protein ß-arrestin. Long PDE4 isoforms thus provide a novel node for cross-talk between the cAMP and p38 MAPK signalling systems at the level of MK2.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Substrate Specificity
10.
Biochem J ; 411(2): 361-9, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18095939

ABSTRACT

We have isolated cDNAs encoding PDE4A8 (phosphodiesterase 4 isoform A8), a new human cAMP-specific PDE4 isoform encoded by the PDE4A gene. PDE4A8 has a novel N-terminal region of 85 amino acids that differs from those of the related 'long' PDE4A4, PDE4A10 and PDE4A11 isoforms. The human PDE4A8 N-terminal region has diverged substantially from the corresponding isoforms in the rat and other mammals, consistent with rapid evolutionary change in this region of the protein. When expressed in COS-7 cells, PDE4A8 localized predominantly in the cytosol, but approx. 20% of the enzyme was associated with membrane fractions. Cytosolic PDE4A8 was exquisitely sensitive to inhibition by the prototypical PDE4 inhibitor rolipram (IC(50) of 11+/-1 nM compared with 1600 nM for PDE4A4), but was less sensitive to inhibition by cilomilast (IC(50) of 101+/-7 nM compared with 61 nM for PDE4A4). PDE4A8 mRNA was found to be expressed predominantly in skeletal muscle and brain, a pattern that differs from the tissue expression of other human PDE4 isoforms and also from that of rat PDE4A8. Immunohistochemical analysis showed that PDE4A8 could be detected in discrete regions of human brain, including the cerebellum, spinal cord and cerebral cortex. The unique tissue distribution of PDE4A8, combined with the evolutionary divergence of its N-terminus, suggest that this isoform may have a specific function in regulating cAMP levels in human skeletal muscle and brain.


Subject(s)
Brain/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Evolution, Molecular , Gene Expression Regulation, Enzymologic , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/isolation & purification , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Genome, Human/genetics , Humans , Molecular Sequence Data , Nucleotides/genetics , Organ Specificity , Phosphorylation , RNA, Messenger/genetics , Rats , Sequence Alignment , Sequence Homology , Time Factors
11.
J Cell Biol ; 177(6): 1119-32, 2007 Jun 18.
Article in English | MEDLINE | ID: mdl-17576803

ABSTRACT

Clearance of fibrin through proteolytic degradation is a critical step of matrix remodeling that contributes to tissue repair in a variety of pathological conditions, such as stroke, atherosclerosis, and pulmonary disease. However, the molecular mechanisms that regulate fibrin deposition are not known. Here, we report that the p75 neurotrophin receptor (p75(NTR)), a TNF receptor superfamily member up-regulated after tissue injury, blocks fibrinolysis by down-regulating the serine protease, tissue plasminogen activator (tPA), and up-regulating plasminogen activator inhibitor-1 (PAI-1). We have discovered a new mechanism in which phosphodiesterase PDE4A4/5 interacts with p75(NTR) to enhance cAMP degradation. The p75(NTR)-dependent down-regulation of cAMP results in a decrease in extracellular proteolytic activity. This mechanism is supported in vivo in p75(NTR)-deficient mice, which show increased proteolysis after sciatic nerve injury and lung fibrosis. Our results reveal a novel pathogenic mechanism by which p75(NTR) regulates degradation of cAMP and perpetuates scar formation after injury.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Fibrosis , Receptor, Nerve Growth Factor/physiology , Tissue Plasminogen Activator/antagonists & inhibitors , Animals , Cicatrix/etiology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Fibrinolysis , Gene Expression Regulation , Mice , Mice, Knockout , Plasminogen Activator Inhibitor 1/genetics , Sciatic Nerve/injuries , Wounds and Injuries
SELECTION OF CITATIONS
SEARCH DETAIL
...