Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Proc Natl Acad Sci U S A ; 115(36): E8469-E8478, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127026

ABSTRACT

Inflammatory responses are terminated by the clearance of dead cells, a process termed efferocytosis. A consequence of efferocytosis is the synthesis of the antiinflammatory mediators TGF-ß, PGE2, and IL-10; however, the efferocytosis of infected cells favors Th17 responses by eliciting the synthesis of TGF-ß, IL-6, and IL-23. Recently, we showed that the efferocytosis of apoptotic Escherichia coli-infected macrophages by dendritic cells triggers PGE2 production in addition to pro-Th17 cytokine expression. We therefore examined the role of PGE2 during Th17 differentiation and intestinal pathology. The efferocytosis of apoptotic E. coli-infected cells by dendritic cells promoted high levels of PGE2, which impaired IL-1R expression via the EP4-PKA pathway in T cells and consequently inhibited Th17 differentiation. The outcome of murine intestinal Citrobacter rodentium infection was dependent on the EP4 receptor. Infected mice treated with EP4 antagonist showed enhanced intestinal defense against C. rodentium compared with infected mice treated with vehicle control. Those results suggest that EP4 signaling during infectious colitis could be targeted as a way to enhance Th17 immunity and host defense.


Subject(s)
Citrobacter rodentium/immunology , Colitis/immunology , Dendritic Cells/immunology , Dinoprostone/immunology , Enterobacteriaceae Infections/immunology , Intestines/immunology , Macrophages/immunology , Animals , Colitis/microbiology , Colitis/pathology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Female , Intestines/microbiology , Macrophages/microbiology , Macrophages/pathology , Mice , Receptors, Prostaglandin E, EP4 Subtype/immunology
2.
J Allergy Clin Immunol ; 146(6): 1387-1396.e13, 2020 12.
Article in English | MEDLINE | ID: mdl-32407837

ABSTRACT

BACKGROUND: Reduced levels of prostaglandin E2 (PGE2) contribute to aspirin-induced hypersensitivity. COX inhibitors are also frequent cofactors in anaphylaxis. Whether alterations in the PGE2 system contribute to anaphylaxis independently of COX inhibitor intake is unclear. OBJECTIVE: Our aim was to test the hypothesis that relative PGE2 deficiency predisposes to anaphylaxis. METHODS: Sera from 48 patients with anaphylaxis and 27 healthy subjects were analyzed for PGE2 levels and correlated against severity; 9α,11ß-PGF2 and PGI2 metabolites were measured for control purposes. PGE2 stabilization by 15-hydroxyprostaglandin dehydrogenase inhibitor or EP2 or EP4 receptor agonists were used in a murine model of passive systemic anaphylaxis. FcεRI-triggered mediator release was determined in bone marrow-derived cultured mast cells (MCs) and human skin-derived MCs. Signaling was studied by Western blot analysis. RESULTS: Patients with anaphylaxis were characterized by markedly reduced PGE2 levels vis-à-vis healthy subjects, whereas prostacyclin metabolite levels were diminished only weakly, and 9α,11ß-PGF2 levels conversely increased. PGE2 was negatively correlated with severity. Lower PGE2 levels and higher susceptibility to anaphylaxis were also found in C57BL/6 mice vis-à-vis in Balb/c mice. Stabilization of PGE2 level by 15-hydroxyprostaglandin dehydrogenase inhibitor protected mice against anaphylaxis. Exogenous PGE2 attenuated bone marrow-derived cultured MC activation through EP2 and EP4 receptors. EP2 and EP4 agonism also curbed FcεRI-mediated degranulation of human MCs. Mechanistically, PGE2 interfered with the phosphorylation of phospholipase C gamma-1 and extracellular signal-regulated kinase. CONCLUSIONS: Homeostatic levels of PGE2 attenuate MC activation via EP2/EP4 and protect against anaphylaxis. Relative deficiency of PGE2 predisposes to anaphylaxis in humans and mice, whereas PGE2 stabilization protects against anaphylactic reactions.


Subject(s)
Anaphylaxis/immunology , Dinoprostone/deficiency , Mast Cells/immunology , Anaphylaxis/pathology , Animals , Dinoprostone/immunology , Disease Susceptibility/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Humans , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Phospholipase C gamma/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Severity of Illness Index
3.
J Allergy Clin Immunol ; 134(5): 1163-74.e16, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24948368

ABSTRACT

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) have recently been implicated in the pathogenesis of asthma, but their regulation in patients with aspirin-intolerant asthma (AIA) remains unclear. OBJECTIVE: We sought to characterize MDSC accumulation and pathogenic functions in allergic airway inflammation mediated by COX-1 deficiency or aspirin treatment in mice. METHODS: Allergic airway inflammation was induced in mice by means of ovalbumin challenge. The distribution and function of MDSCs in mice were analyzed by using flow cytometry and pharmacologic/gene manipulation approaches. RESULTS: CD11b(+)Gr1(high)Ly6G(+)Ly6C(int) MDSCs (polymorphonuclear MDSCs [PMN-MDSCs]) recruited to the lungs are negatively correlated with airway inflammation in allergen-challenged mice. Aspirin-treated and COX-1 knockout (KO) mice showed significantly lower accumulation of PMN-MDSCs in the inflamed lung and immune organs accompanied by increased TH2 airway responses. The TH2-suppressive function of PMN-MDSCs was notably impaired by COX-1 deletion or inhibition, predominantly through downregulation of arginase-1. COX-1-derived prostaglandin E2 promoted PMN-MDSC generation in bone marrow through E prostanoid 2 and 4 receptors (EP2 and EP4), whereas the impaired arginase-1 expression in PMN-MDSCs in COX-1 KO mice was mediated by dysregulation of the prostaglandin E2/EP4/cyclic AMP/protein kinase A pathway. EP4 agonist administration alleviated allergy-induced airway hyperresponsiveness in COX-1 KO mice. Moreover, the immunosuppressive function of PMN-MDSCs from patients with AIA was dramatically decreased compared with that from patients with aspirin-tolerant asthma. CONCLUSION: The immunosuppressive activity of PMN-MDSCs was diminished in both allergen-challenged COX-1 KO mice and patients with AIA, probably through an EP4-mediated signaling pathway, indicating that activation of PMN-MDSCs might be a promising therapeutic strategy for asthma, particularly AIA.


Subject(s)
Asthma, Aspirin-Induced/immunology , Immune Tolerance , Myeloid Cells/immunology , Signal Transduction/immunology , Allergens/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Arginase/genetics , Arginase/immunology , Aspirin/adverse effects , Aspirin/pharmacology , Asthma, Aspirin-Induced/genetics , Asthma, Aspirin-Induced/pathology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/immunology , Cyclooxygenase 1/genetics , Cyclooxygenase 1/immunology , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Myeloid Cells/pathology , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Th2 Cells/immunology , Th2 Cells/pathology
4.
J Immunol ; 189(10): 4777-86, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23066152

ABSTRACT

Seminal plasma is not just a carrier for spermatozoa. It contains high concentrations of cytokines, chemokines, and other biological compounds that are able to exert potent effects on the immune system of the receptive partner. Previous studies have shown that semen induces an acute inflammatory response at the female genital mucosa after coitus. Moreover, it induces regulatory mechanisms that allow the fetus (a semiallograft) to grow and develop in the uterus. The mechanisms underlying these regulatory mechanisms, however, are poorly understood. In this study, we show that seminal plasma redirects the differentiation of human dendritic cells (DCs) toward a regulatory profile. DCs differentiated from human monocytes in the presence of high dilutions of seminal plasma did not express CD1a but showed high levels of CD14. They were unable to develop a fully mature phenotype in response to LPS, TNF-α, CD40L, Pam2CSK4 (TLR2/6 agonist), or Pam3CSK4 (TLR1/2 agonist). Upon activation, they produced low amounts of the inflammatory cytokines IL-12p70, IL-1ß, TNF-α, and IL-6, but expressed a high ability to produce IL-10 and TGF-ß. Inhibition of the PG receptors E-prostanoid receptors 2 and 4 prevented the tolerogenic effect induced by seminal plasma on the phenotype and function of DCs, suggesting that E-series PGs play a major role. By promoting a tolerogenic profile in DCs, seminal plasma might favor fertility, but might also compromise the capacity of the receptive partner to mount an effective immune response against sexually transmitted pathogens.


Subject(s)
Cell Differentiation/physiology , Dendritic Cells/immunology , Immune Tolerance/physiology , Monocytes/immunology , Semen/immunology , Adult , Antigens, CD1/immunology , Cell Differentiation/drug effects , Cytokines/immunology , Dendritic Cells/cytology , Female , Humans , Immune Tolerance/drug effects , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , Male , Monocytes/cytology , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/immunology
5.
Proc Natl Acad Sci U S A ; 108(16): 6668-73, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21460251

ABSTRACT

UV radiation induces systemic immunosuppression. Because nonsteroidal anti-inflammatory drugs suppress UV-induced immunosuppression, prostanoids have been suspected as a crucial mediator of this UV effect. However, the identity of the prostanoid involved and its mechanism of action remain unclear. Here, we addressed this issue by subjecting mice deficient in each prostanoid receptor individually or mice treated with a subtype-specific antagonist to UV irradiation. Mice treated with an antagonist for prostaglandin E receptor subtype 4 (EP4), but not those deficient in other prostanoid receptors, show impaired UV-induced immunosuppression, whereas administration of an EP4 agonist rescues the impairment of the UV-induced immunosuppression in indomethacin-treated mice. The EP4 antagonist treatment suppresses an increase in the number of CD4(+)/forkhead box P3-positive (Foxp3(+)) regulatory T cells (Treg cells) in the peripheral lymph nodes (LNs) and dendritic cells expressing DEC205 in the LNs and the skin after UV irradiation. Furthermore, the EP4 antagonist treatment down-regulates UV-induced expression of receptor activator of NF-κB ligand (RANKL) in skin keratinocytes. Finally, administration of anti-RANKL antibody abolishes the restoration of UV-induced immunosuppression by EP4 agonism in indomethacin-treated mice. Thus, prostaglandin E(2) (PGE(2))-EP4 signaling mediates UV-induced immunosuppression by elevating the number of Treg cells through regulation of RANKL expression in the epidermis.


Subject(s)
Dinoprostone/immunology , Immune Tolerance/radiation effects , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/immunology , Ultraviolet Rays/adverse effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dinoprostone/genetics , Dinoprostone/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Gene Expression Regulation/radiation effects , Immune Tolerance/drug effects , Immune Tolerance/genetics , Immune Tolerance/immunology , Indomethacin/pharmacology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Transgenic , RANK Ligand/biosynthesis , RANK Ligand/genetics , RANK Ligand/immunology , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
6.
Eur J Immunol ; 42(4): 912-23, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22531917

ABSTRACT

Gastrointestinal inflammation is mediated by the pro-inflammatory mediators interleukin-8 (IL-8) and prostaglandin E(2) (PGE(2) ). PGE(2) binding and coupling through EP2/4 receptor subtypes on colonic epithelial cells stimulates cyclic adenosine monophosphate (cAMP) and IL-8 production. Here we determined the mechanisms whereby PGE(2) regu-lates IL-8 in Caco2 colonic epithelial cells and in cells over-expressing the EP2/4 receptors (EP2S/EP4S). PGE(2) coupling through EP2 activated the transcription factor inducible cAMP early repressor (ICER), whereas coupling through EP4 receptors activated the cyclic AMP-responsive element-binding protein (CREB). Activation of CREB in Caco2/EP2S was protein kinase A (PKA) dependent, whereas in EP4S cells, activation of CREB occurred through the PKA and phosphatidylinositol 3-kinase pathways. Since ICER lacks the transactivation domain, it functions as a transcription repressor as opposed to CREB. PGE(2) coupling through EP2/4 receptors can therefore acts in an opposing manner to either decrease (EP2) or promote IL-8 expression by recruiting CREB-binding protein (CBP) (EP4), which formed a multiprotein IL-8 enhanceosome. A novel half CRE (167CRE) and a composite NFAT1-AP1-like site in the IL-8 promoter participated in binding and complex formation as confirmed by mutagenesis and expression studies. These data unravel the mechanisms by which expression of IL-8 is controlled by different signalling pathways that are activated by PGE(2) but acting through different EP receptors.


Subject(s)
Colon/immunology , Dinoprostone/immunology , Epithelial Cells/immunology , Gene Expression Regulation/immunology , Interleukin-8/immunology , Intestinal Mucosa/immunology , Caco-2 Cells , Colon/cytology , Colon/metabolism , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/immunology , Cyclic AMP Response Element Modulator/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/genetics , Dinoprostone/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation/genetics , Humans , Interleukin-8/biosynthesis , Interleukin-8/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Response Elements/genetics , Response Elements/immunology , Signal Transduction/genetics , Signal Transduction/immunology
7.
Prostaglandins Other Lipid Mediat ; 100-101: 30-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23237743

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis (MS), which is a T cell-mediated autoimmune disease of the central nervous system (CNS). Emerging evidence indicates that both prostaglandin E (PGE) and adenosine play important roles in immune inflammation although the mechanism remains unclear. In the study, we examined individual and combined effect of PGE and adenosine during EAE development. The results showed that both PGE and adenosine could inhibit EAE progression and they in combination showed substantially higher inhibition than each modality used alone. On the other hand, using specific agonists or antagonists for PGE and adenosine receptors indicated that the suppression of EAE development was mainly mediated by EP4 and A receptors. Furthermore, combined PGE and adenosine treatment significantly suppressed the production of IFN-γ and IL-17 via EP4 and A receptors. Taken together, PGE and adenosine in combination could protect EAE mouse from serious EAE through limiting the over-reactive effects of T cells via EP4 and A receptors.


Subject(s)
Adenosine/pharmacology , Dinoprostone/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Adenosine/therapeutic use , Analysis of Variance , Animals , Dinoprostone/therapeutic use , Drug Synergism , Drug Therapy, Combination , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Receptor, Adenosine A2A/immunology , Receptor, Adenosine A2A/metabolism , Receptors, Prostaglandin E, EP4 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors
8.
Cell Immunol ; 273(2): 109-14, 2012.
Article in English | MEDLINE | ID: mdl-22321156

ABSTRACT

Follicular dendritic cells (FDCs) are an essential cellular component of the germinal center (GC) and are believed to exert regulatory effects on the various stages of GC reactions. According to our previous reports, human FDCs express prostacyclin synthase, and prostacyclin analogues augment adhesion and co-stimulatory molecules on the surface of activated B cells. These findings prompted us to investigate whether FDCs would contribute to the antigen-presenting capability of B cells by using the well-established FDC-like cells, HK cells, and tonsillar B cells. Our results show that HK cells significantly enhance the expression levels of CD54, CD80, and CD86 on the surface of activated B cells. The enhancing effect of HK cells on CD86 is impeded by indomethacin and an EP4 antagonist, implying that a certain prostaglandin is mediating the up-regulation. Prostacyclin indeed recapitulates the enhancing effect on CD86, which is inhibited by EP4 as well as IP antagonists. B cells co-cultured with HK cells exhibit an augmented APC activity, which is inhibited by CD86 neutralization. These results reveal another unrecognized function of human FDC.


Subject(s)
B-Lymphocytes/immunology , B7-2 Antigen/immunology , Dendritic Cells, Follicular/immunology , Immunity, Innate , Antigen Presentation , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B7-2 Antigen/antagonists & inhibitors , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Cell Adhesion/immunology , Cell Communication/genetics , Cell Communication/immunology , Coculture Techniques , Cytochrome P-450 Enzyme System/immunology , Cytochrome P-450 Enzyme System/metabolism , Dendritic Cells, Follicular/cytology , Dendritic Cells, Follicular/drug effects , Epoprostenol/immunology , Epoprostenol/metabolism , Flow Cytometry , Gene Expression , Humans , Indomethacin/pharmacology , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/metabolism , Lymphocyte Activation , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Primary Cell Culture , Prostaglandin Antagonists/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
9.
Postepy Hig Med Dosw (Online) ; 66: 287-94, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22706114

ABSTRACT

Prevention and treatment of pathological inflammatory processes requires application of various classes of immune suppressors, such as calcineurin inhibitors, steroids and non-steroid inhibitors of prostaglandin synthesis. However, each type of these immune suppressors causes less or more serious adverse side-effects. Exploration of the role played by prostanoids in the immune response and identification of functionally distinct prostaglandin E receptors (EP1-EP4) opened new perspectives in therapy of inflammation, autoimmunity and prevention of graft rejection. The EP4 receptor appeared to be an attractive target to affect manifestations of various pathological states by application of either agonists or antagonists of the receptor. This article presents a short overview of experimental approaches aimed at manipulation of signaling via EP2 and EP4 receptors that could have therapeutic utility.


Subject(s)
Inflammation/drug therapy , Inflammation/immunology , Receptors, Prostaglandin E, EP2 Subtype/drug effects , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/drug effects , Receptors, Prostaglandin E, EP4 Subtype/immunology , Animals , Arthritis/drug therapy , Arthritis/immunology , Autoimmunity/drug effects , Autoimmunity/immunology , Dinoprostone/pharmacology , Gastroenteritis/drug therapy , Gastroenteritis/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Prostaglandins , Reperfusion Injury/immunology , Reperfusion Injury/prevention & control
10.
Cancer Immunol Immunother ; 60(11): 1577-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21681369

ABSTRACT

The COX-2 product prostaglandin E(2) (PGE(2)) contributes to the high metastatic capacity of breast tumors. Our published data indicate that inhibiting either PGE(2) production or PGE(2)-mediated signaling through the PGE(2) receptor EP4 reduces metastasis by a mechanism that requires natural killer (NK) cells. It is known that NK cell function is compromised by PGE(2), but very little is known about the mechanism by which PGE(2) affects NK effector activity. We now report the direct effects of PGE(2) on the NK cell. Endogenous murine splenic NK cells express all four PGE(2) receptors (EP1-4). We examined the role of EP receptors in three NK cell functions: migration, cytotoxicity, and cytokine release. Like PGE(2), the EP4 agonist PGE(1)-OH blocked NK cell migration to FBS and to four chemokines (ITAC, MIP-1α, SDF-1α, and CCL21). The EP2 agonist, Butaprost, inhibited migration to specific chemokines but not in response to FBS. In contrast to the inhibitory actions of PGE(2), the EP1/EP3 agonist Sulprostone increased migration. Unlike the opposing effects of EP4 vs. EP1/EP3 on migration, agonists of each EP receptor were uniformly inhibiting to NK-mediated cytotoxicity. The EP4 agonist, PGE(1)-OH, inhibited IFNγ production from NK cells. Agonists for EP1, EP2, and EP3 were not as effective at inhibiting IFNγ. Agonists of EP1, EP2, and EP4 all inhibited TNFα; EP4 agonists were the most potent. Thus, the EP4 receptor consistently contributed to loss of function. These results, taken together, support a mechanism whereby inhibiting PGE(2) production or preventing signaling through the EP4 receptor may prevent suppression of NK functions that are critical to the control of breast cancer metastasis.


Subject(s)
Dinoprostone/immunology , Killer Cells, Natural/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Animals , Cell Line, Tumor , Dinoprostone/pharmacology , Female , Lymphoma, T-Cell/immunology , Mice , Mice, Inbred BALB C
11.
Chem Pharm Bull (Tokyo) ; 59(12): 1523-34, 2011.
Article in English | MEDLINE | ID: mdl-22130375

ABSTRACT

Analogs 8-aza-16-aryl prostaglandin E(1) (PGE(1)) and 8-aza-5-thia-16-arylPGE(1) were synthesized and evaluated with respect to their subtype receptor affinity and EP4 agonist activity for the purposes of identifying subtype-selective EP4 agonists that demonstrate oral efficacy. Using an inhibition assay of lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF)-α production in rats, representative compounds were evaluated for their pharmacokinetic profiles and in vivo efficacy. Structure-activity relationships (SARs) were characterized and presented. Of the compounds tested, several demonstrated better oral exposure and/or in vivo efficacy compared with the previously reported analog 2a.


Subject(s)
Alprostadil/analogs & derivatives , Prostaglandins E, Synthetic/chemistry , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/agonists , Administration, Oral , Alprostadil/administration & dosage , Alprostadil/chemical synthesis , Alprostadil/chemistry , Alprostadil/pharmacokinetics , Alprostadil/pharmacology , Animals , Humans , Lipopolysaccharides/immunology , Prostaglandins E, Synthetic/administration & dosage , Prostaglandins E, Synthetic/chemical synthesis , Prostaglandins E, Synthetic/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP4 Subtype/immunology , Tumor Necrosis Factor-alpha/immunology
12.
Front Immunol ; 11: 1224, 2020.
Article in English | MEDLINE | ID: mdl-32636842

ABSTRACT

Increased serum IgE level is one of the features of allergic asthma. It is reported that IgE production can be enhanced by E-prostanoid 2 (EP2) receptor of prostaglandin E2 (PGE2); however, whether E-prostanoid 4 (EP4) receptor (encoded by Ptger4) has a unique or redundant role is still unclear. Here, we demonstrated the mice with B cell-specific deletion of the EP4 receptor (Ptger4fl/flMb1cre+/-) showed their serum levels of IgE were markedly increased. A much more severe airway allergic inflammation was observed in the absence of EP4 signal using the OVA-induced asthma model. Mechanistic studies demonstrated that the transcription levels of AID, GLTε, and PSTε in EP4-deficient B cells were found to be significantly increased, implying an enhanced IgE class switch. In addition, we saw higher levels of phosphorylated STAT6, a vital factor for IgE class switch. Biochemical analyses indicated that inhibitory effect of EP4 signal on IgE depended on the activation of the PI3K-AKT pathway. Further downstream, PPARγ expression was up-regulated. Independent of its activity as a transcription factor, PPARγ here primarily functioned as an E3 ubiquitin-ligase, which bound the phosphorylated STAT6 to initiate its degradation. In support of PPARγ as a key mediator downstream of the EP4 signal, PPARγ agonist induced the down-regulation of phospho-STAT6, whereas its antagonist was able to rescue the EP4-mediated inhibition of STAT6 activation and IgE production. Thus, our findings highlight a role for the PGE2-EP4-AKT-PPARγ-STAT6 signaling in IgE response, highlighting the therapeutic potential of combined application of EP4 and PPARγ agonists in asthma.


Subject(s)
Asthma/metabolism , Dinoprostone/metabolism , Immunoglobulin Class Switching/immunology , Immunoglobulin E/biosynthesis , PPAR gamma/metabolism , Animals , Asthma/immunology , Dinoprostone/immunology , Immunoglobulin E/immunology , Mice , Mice, Inbred C57BL , PPAR gamma/immunology , Protein Stability , Receptors, Prostaglandin E, EP4 Subtype/deficiency , Receptors, Prostaglandin E, EP4 Subtype/immunology , STAT6 Transcription Factor/metabolism , Signal Transduction/immunology
13.
Sci Rep ; 8(1): 13351, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30190508

ABSTRACT

Cardiosphere-derived cells (CDCs) are a cardiac progenitor cell population, which have been shown to possess cardiac regenerative properties and can improve heart function in a variety of cardiac diseases. Studies in large animal models have predominantly focussed on using autologous cells for safety, however allogeneic cell banks would allow for a practical, cost-effective and efficient use in a clinical setting. The aim of this work was to determine the immunomodulatory status of these cells using CDCs and lymphocytes from 5 dogs. CDCs expressed MHC I but not MHC II molecules and in mixed lymphocyte reactions demonstrated a lack of lymphocyte proliferation in response to MHC-mismatched CDCs. Furthermore, MHC-mismatched CDCs suppressed lymphocyte proliferation and activation in response to Concanavalin A. Transwell experiments demonstrated that this was predominantly due to direct cell-cell contact in addition to soluble mediators whereby CDCs produced high levels of PGE2 under inflammatory conditions. This led to down-regulation of CD25 expression on lymphocytes via the EP4 receptor. Blocking prostaglandin synthesis restored both, proliferation and activation (measured via CD25 expression) of stimulated lymphocytes. We demonstrated for the first time in a large animal model that CDCs inhibit proliferation in allo-reactive lymphocytes and have potent immunosuppressive activity mediated via PGE2.


Subject(s)
Dinoprostone/immunology , Immune Tolerance , Lymphocytes/immunology , Myocardium/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Stem Cells/immunology , Animals , Cell Communication/immunology , Cell Proliferation , Dogs , Histocompatibility Antigens Class I/immunology , Lymphocytes/cytology , Myocardium/cytology , Stem Cells/cytology
14.
Front Immunol ; 9: 2954, 2018.
Article in English | MEDLINE | ID: mdl-30619314

ABSTRACT

PGE2 is a lipid mediator of the initiation and resolution phases of inflammation, as well as a regulator of immune system responses to inflammatory events. PGE2 is produced and sensed by T cells, and autocrine or paracrine PGE2 can affect T cell phenotype and function. In this study, we use a T cell-dependent model of colitis to evaluate the role of PGE2 on pathological outcome and T-cell phenotypes. CD4+ T effector cells either deficient in mPGES-1 or the PGE2 receptor EP4 are less colitogenic. Absence of T cell autocrine mPGES1-dependent PGE2 reduces colitogenicity in association with an increase in CD4+RORγt+ cells in the lamina propria. In contrast, recipient mice deficient in mPGES-1 exhibit more severe colitis that corresponds with a reduced capacity to generate FoxP3+ T cells, especially in mesenteric lymph nodes. Thus, our research defines how mPGES-1-driven production of PGE2 by different cell types in distinct intestinal locations impacts T cell function during colitis. We conclude that PGE2 has profound effects on T cell phenotype that are dependent on the microenvironment.


Subject(s)
Colitis/immunology , Dinoprostone/immunology , Prostaglandin-E Synthases/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis/genetics , Colitis/metabolism , Dinoprostone/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
15.
JCI Insight ; 3(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29415894

ABSTRACT

Type I IFN (IFN-I) dysregulation contributes to type 1 diabetes (T1D) development, and although increased IFN-I signals are pathogenic at the initiation of autoimmune diabetes, IFN-I dysregulation at later pathogenic stages more relevant for therapeutic intervention is not well understood. We discovered that 5 key antigen-presenting cell subsets from adult prediabetic NOD mice have reduced responsiveness to IFN-I that is dominated by a decrease in the tonic-sensitive subset of IFN-I response genes. Blockade of IFNAR1 in prediabetic NOD mice accelerated diabetes and increased Th1 responses. Therefore, IFN-I responses shift from pathogenic to protective as autoimmunity progresses, consistent with chronic IFN-I exposure. In contrast, IL-1-associated inflammatory pathways were elevated in prediabetic mice. These changes correlated with human T1D onset-associated gene expression. Prostaglandin E2 (PGE2) and prostaglandin receptor 4 (PTGER4), a receptor for PGE2 that mediates both inflammatory and regulatory eicosanoid signaling, were higher in NOD mice and drive innate immune dysregulation. Treating prediabetic NOD mice with a PTGER4 antagonist restored IFNAR signaling, decreased IL-1 signaling, and decreased infiltration of leukocytes into the islets. Therefore, innate cytokine alterations contribute to both T1D-associated inflammation and autoimmune pathogenesis. Modulating innate immune balance via signals such as PTGER4 may contribute to treatments for autoimmunity.


Subject(s)
Autoimmunity/drug effects , Diabetes Mellitus, Type 1/immunology , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Sulfonylurea Compounds/administration & dosage , Th1 Cells/immunology , Administration, Oral , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Diabetes Mellitus, Type 1/drug therapy , Dinoprostone/immunology , Dinoprostone/metabolism , Disease Models, Animal , Female , Humans , Interleukin-1/immunology , Interleukin-1/metabolism , Mice , Mice, Inbred NOD , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Receptors, Prostaglandin E, EP4 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Th1 Cells/drug effects , Th1 Cells/metabolism
16.
Diabetes ; 66(2): 494-504, 2017 02.
Article in English | MEDLINE | ID: mdl-27815317

ABSTRACT

Diabetic nephropathy (DN) is characterized by increased macrophage infiltration, and proinflammatory M1 macrophages contribute to development of DN. Previous studies by us and others have reported that macrophage cyclooxygenase-2 (COX-2) plays a role in polarization and maintenance of a macrophage tissue-reparative M2 phenotype. We examined the effects of macrophage COX-2 on development of DN in type 1 diabetes. Cultured macrophages with COX-2 deletion exhibited an M1 phenotype, as demonstrated by higher inducible nitric oxide synthase and nuclear factor-κB levels but lower interleukin-4 receptor-α levels. Compared with corresponding wild-type diabetic mice, mice with COX-2 deletion in hematopoietic cells (COX-2 knockout bone marrow transplantation) or macrophages (CD11b-Cre COX2f/f) developed severe DN, as indicated by increased albuminuria, fibrosis, and renal infiltration of T cells, neutrophils, and macrophages. Although diabetic kidneys with macrophage COX-2 deletion had more macrophage infiltration, they had fewer renal M2 macrophages. Diabetic kidneys with macrophage COX-2 deletion also had increased endoplasmic reticulum stress and decreased number of podocytes. Similar results were found in diabetic mice with macrophage PGE2 receptor subtype 4 deletion. In summary, these studies have demonstrated an important but unexpected role for macrophage COX-2/prostaglandin E2/PGE2 receptor subtype 4 signaling to lessen progression of diabetic kidney disease, unlike the pathogenic effects of increased COX-2 expression in intrinsic renal cells.


Subject(s)
Cyclooxygenase 2/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Diabetic Nephropathies/immunology , Macrophages/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Albuminuria , Animals , Cells, Cultured , Cyclooxygenase 2/genetics , Fibrosis , Immunoblotting , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Macrophages/metabolism , Male , Mice , Mice, Knockout , NF-kappa B/immunology , Neutrophil Infiltration/immunology , Neutrophils , Nitric Oxide Synthase Type II/immunology , Real-Time Polymerase Chain Reaction , Receptors, Prostaglandin E/immunology , Signal Transduction , T-Lymphocytes/immunology
17.
Cell Mol Immunol ; 13(2): 240-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26189370

ABSTRACT

While there is mounting evidence that interleukin (IL)-23-IL-17 axis plays a critical role in the pathogenesis of various autoimmune diseases, much remains to be elucidated on how IL-23 is induced in the pathological processes. IL-23 is a heterodimer composed of p19 and p40, the latter being shared with IL-12. We previously reported that prostaglandin (PG) E2 promotes CD40-mediated induction of Il23a (p19) expression through its E receptor subtype 4 (EP4) receptor in splenic dendritic cells (DCs). Here, we have analyzed signaling pathways regulating Il23a induction in the cross talk between EP4 and CD40 in bone marrow-derived DCs. We found that PGE2 synergistically induced Il23a transcription with CD40 signaling. An EP4 agonist, but not agonists of EP1, EP2, or EP3, reproduced this action. Stimulation of CD40 with an agonist antibody evoked biphasic induction of Il23a expression, with the early phase peaking at 1 h and the late phase peaking at 12 h and lasting up to 36 h after stimulation, whereas induction by lipopolysaccharide or tumor necrosis factor-α was transient. The early phase induction by CD40 stimulation was absent in DCs derived from Nfkb1-deficient mice, and the late phase induction was eliminated by RNA interference of nuclear factor-kappa B (NF-κB) p100 subunit. Further, cAMP response element-binding protein (CREB) depletion completely eliminated the induction of Il23a by CD40 stimulation. The addition of the EP4 agonist amplified the induction in both phases through the cAMP-protein kinase A (PKA) pathway. These results suggest that Il23a expression in DCs is synergistically triggered by the PG E2-EP4-cAMP-PKA pathway and canonical/non-canonical NF-κB pathways and CREB activated by CD40 stimulation.


Subject(s)
CD40 Antigens/immunology , Dendritic Cells/immunology , Dinoprostone/immunology , Interleukin-23 Subunit p19/immunology , NF-kappa B p50 Subunit/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Signal Transduction/immunology , Animals , CD40 Antigens/genetics , Dinoprostone/genetics , Female , Interleukin-23 Subunit p19/genetics , Mice , Mice, Mutant Strains , NF-kappa B p50 Subunit/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Signal Transduction/genetics , Transcription, Genetic/immunology
18.
Science ; 351(6279): 1333-8, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26989254

ABSTRACT

Systemic inflammation, which results from the massive release of proinflammatory molecules into the circulatory system, is a major risk factor for severe illness, but the precise mechanisms underlying its control are not fully understood. We observed that prostaglandin E2 (PGE2), through its receptor EP4, is down-regulated in human systemic inflammatory disease. Mice with reduced PGE2 synthesis develop systemic inflammation, associated with translocation of gut bacteria, which can be prevented by treatment with EP4 agonists. Mechanistically, we demonstrate that PGE2-EP4 signaling acts directly on type 3 innate lymphoid cells (ILCs), promoting their homeostasis and driving them to produce interleukin-22 (IL-22). Disruption of the ILC-IL-22 axis impairs PGE2-mediated inhibition of systemic inflammation. Hence, the ILC-IL-22 axis is essential in protecting against gut barrier dysfunction, enabling PGE2-EP4 signaling to impede systemic inflammation.


Subject(s)
Dinoprostone/immunology , Inflammation/immunology , Interleukins/immunology , Intestines/immunology , Lymphocytes/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Gene Expression , Humans , Immunity, Innate , Inflammation/drug therapy , Inflammation/microbiology , Intestines/microbiology , Mice , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/genetics , Signal Transduction , Interleukin-22
19.
PLoS One ; 11(1): e0147721, 2016.
Article in English | MEDLINE | ID: mdl-26824354

ABSTRACT

Inflammatory activation of microglia and ß amyloid (Aß) deposition are considered to work both independently and synergistically to contribute to the increased risk of Alzheimer's disease (AD). Recent studies indicate that long-term use of phenolic compounds provides protection against AD, primarily due to their anti-inflammatory actions. We previously suggested that phenolic compound curcumin ameliorated phagocytosis possibly through its anti-inflammatory effects rather than direct regulation of phagocytic function in electromagnetic field-exposed N9 microglial cells (N9 cells). Here, we explored the prostaglandin-E2 (PGE2)-related signaling pathway that involved in curcumin-mediated phagocytosis in fibrillar ß-amyloid peptide (1-42) (fAß42)-stimulated N9 cells. Treatment with fAß42 increased phagocytosis of fluorescent-labeled latex beads in N9 cells. This increase was attenuated in a dose-dependent manner by endogenous and exogenous PGE2, as well as a selective EP2 or protein kinase A (PKA) agonist, but not by an EP4 agonist. We also found that an antagonist of EP2, but not EP4, abolished the reduction effect of PGE2 on fAß42-induced microglial phagocytosis. Additionally, the increased expression of endogenous PGE2, EP2, and cyclic adenosine monophosphate (AMP), and activation of vasodilator-stimulated phosphoprotein, cyclic AMP responsive element-binding protein, and PKA were depressed by curcumin administration. This reduction led to the amelioration of the phagocytic abilities of PGE2-stimulated N9 cells. Taken together, these data suggested that curcumin restored the attenuating effect of PGE2 on fAß42-induced microglial phagocytosis via a signaling mechanism involving EP2 and PKA. Moreover, due to its immune modulatory effects, curcumin may be a promising pharmacological candidate for neurodegenerative diseases.


Subject(s)
Amyloid beta-Peptides/immunology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Curcumin/pharmacology , Dinoprostone/immunology , Microglia/drug effects , Microglia/immunology , Peptide Fragments/immunology , Phagocytosis/drug effects , Animals , Cyclic AMP/immunology , Cyclic AMP-Dependent Protein Kinases/immunology , Mice , Microglia/cytology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Signal Transduction/drug effects
20.
Nat Commun ; 7: 10760, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26907476

ABSTRACT

Tityus serrulatus sting causes thousands of deaths annually worldwide. T. serrulatus-envenomed victims exhibit local or systemic reaction that culminates in pulmonary oedema, potentially leading to death. However, the molecular mechanisms underlying T. serrulatus venom (TsV) activity remain unknown. Here we show that TsV triggers NLRP3 inflammasome activation via K(+) efflux. Mechanistically, TsV triggers lung-resident cells to release PGE2, which induces IL-1ß production via E prostanoid receptor 2/4-cAMP-PKA-NFκB-dependent mechanisms. IL-1ß/IL-1R actions account for oedema and neutrophil recruitment to the lungs, leading to TsV-induced mortality. Inflammasome activation triggers LTB4 production and further PGE2 via IL-1ß/IL-1R signalling. Activation of LTB4-BLT1/2 pathway decreases cAMP generation, controlling TsV-induced inflammation. Exogenous administration confirms LTB4 anti-inflammatory activity and abrogates TsV-induced mortality. These results suggest that the balance between LTB4 and PGE2 determines the amount of IL-1ß inflammasome-dependent release and the outcome of envenomation. We suggest COX1/2 inhibition as an effective therapeutic intervention for scorpion envenomation.


Subject(s)
Carrier Proteins/genetics , Dinoprostone/pharmacology , Interleukin-1beta/drug effects , Leukotriene B4/pharmacology , Macrophages, Peritoneal/drug effects , Scorpion Stings/immunology , Scorpion Venoms/pharmacology , Animals , Arachidonate 5-Lipoxygenase/genetics , Blotting, Western , Carrier Proteins/immunology , Celecoxib/pharmacology , Cyclic AMP/immunology , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/immunology , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/immunology , In Vitro Techniques , Indoles/pharmacology , Indomethacin/pharmacology , Inflammasomes/immunology , Interleukin-1beta/immunology , Leukotriene B4/immunology , Lipoxygenase Inhibitors/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout , NF-kappa B/drug effects , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphoproteins , Prostaglandin Antagonists/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/drug effects , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/drug effects , Receptors, Prostaglandin E, EP4 Subtype/immunology , Reverse Transcriptase Polymerase Chain Reaction , Scorpion Stings/mortality , Scorpions , Xanthones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL