Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Neurosci ; 41(5): 1105-1117, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33293358

ABSTRACT

A multidimensional inflammatory response ensues after status epilepticus (SE), driven partly by cyclooxygenase-2-mediated activation of prostaglandin EP2 receptors. The inflammatory response is typified by astrocytosis, microgliosis, erosion of the blood-brain barrier (BBB), formation of inflammatory cytokines, and brain infiltration of blood-borne monocytes. Our previous studies have shown that inhibition of monocyte brain invasion or systemic administration of an EP2 receptor antagonist relieves multiple deleterious consequences of SE. Here we identify those effects of EP2 antagonism that are reproduced by conditional ablation of EP2 receptors in immune myeloid cells and show that systemic EP2 antagonism blocks monocyte brain entry in male mice. The induction of hippocampal IL-6 after pilocarpine SE was nearly abolished in EP2 conditional KO mice. Serum albumin levels in the cortex, a measure of BBB breakdown, were significantly higher after SE in EP2-sufficient mice but not in EP2 conditional KOs. EP2 deficiency in innate immune cells accelerated the recovery from sickness behaviors following SE. Surprisingly, neurodegeneration was not alleviated in myeloid conditional KOs. Systemic EP2 antagonism prevented monocyte brain infiltration and provided broader rescue of SE-induced effects than myeloid EP2 ablation, including neuroprotection and broader suppression of inflammatory mediators. Reporter expression indicated that the cellular target of CD11b-driven Cre was circulating myeloid cells but, unexpectedly, not microglia. These findings indicate that activation of EP2 receptors on immune myeloid cells drives substantial deficits in behavior and disrupts the BBB after SE. The benefits of systemic EP2 antagonism can be attributed, in part, to blocking brain recruitment of blood-borne monocytes.SIGNIFICANCE STATEMENT Unabated seizures reduce quality of life, promote the development of epilepsy, and can be fatal. We previously identified activation of prostaglandin EP2 receptors as a driver of undesirable consequences of seizures. However, the relevant EP2-expressing cell types remain unclear. Here we identify peripheral innate immune cells as a driver of the EP2-related negative consequences of seizures. Removal of EP2 from peripheral immune cells was beneficial, abolishing production of a key inflammatory cytokine, accelerating weight regain, and limiting behavioral deficits. These findings provide evidence that EP2 engagement on peripheral immune and brain endothelia contributes to the deleterious effects of SE, and will assist in the development of beneficial therapies to enhance quality of life in individuals who suffer prolonged seizures.


Subject(s)
Immunity, Innate/physiology , Myeloid Cells/metabolism , Receptors, Prostaglandin E, EP2 Subtype/biosynthesis , Status Epilepticus/metabolism , Animals , Flow Cytometry/methods , Hippocampus/cytology , Hippocampus/immunology , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/immunology , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/immunology , Status Epilepticus/genetics , Status Epilepticus/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.
Mol Pharm ; 15(12): 5809-5817, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30398879

ABSTRACT

The prostaglandin E2 receptor, EP2, plays an important role in physiology and in a variety of pathological conditions. Studies indicate that EP2 is pro-inflammatory in chronic peripheral and central nervous system disease and cancer models. Thus, targeting the EP2 receptor with small molecules could be a therapeutic strategy for treating inflammatory diseases and cancer. We recently reported a novel class of competitive antagonists of the EP2 receptor. However, earlier leads displayed low selectivity against the DP1 prostanoid receptor, moderate plasma half-life, and low aqueous solubility, which renders them suboptimal for testing in animal models of disease. We now report a novel compound TG8-69, which has suitable drug-like properties. We present synthesis, lead-optimization studies, pharmacological characterization, and anti-inflammatory properties of this compound that support its use in chronic peripheral inflammatory diseases, including rheumatoid arthritis, endometriosis, and cancer, in which EP2 appears to play a pathogenic role.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation Mediators/metabolism , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Up-Regulation/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Cell Line , Dinoprostone/immunology , Dinoprostone/metabolism , Drug Evaluation, Preclinical , Endometriosis/drug therapy , Endometriosis/immunology , Female , Half-Life , Inflammation Mediators/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/immunology , Rats , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Solubility , Up-Regulation/immunology , Water/chemistry
4.
Nat Immunol ; 19(12): 1391-1402, 2018 12.
Article in English | MEDLINE | ID: mdl-30374130

ABSTRACT

Foxp3+ regulatory T cells (Treg cells) are the central component of peripheral immune tolerance. Whereas a dysregulated Treg cytokine signature has been observed in autoimmune diseases, the regulatory mechanisms underlying pro- and anti-inflammatory cytokine production are elusive. Here, we identify an imbalance between the cytokines IFN-γ and IL-10 as a shared Treg signature present in patients with multiple sclerosis and under high-salt conditions. RNA-sequencing analysis on human Treg subpopulations revealed ß-catenin as a key regulator of IFN-γ and IL-10 expression. The activated ß-catenin signature was enriched in human IFN-γ+ Treg cells, as confirmed in vivo with Treg-specific ß-catenin-stabilized mice exhibiting lethal autoimmunity with a dysfunctional Treg phenotype. Moreover, we identified prostaglandin E receptor 2 (PTGER2) as a regulator of IFN-γ and IL-10 production under a high-salt environment, with skewed activation of the ß-catenin-SGK1-Foxo axis. Our findings reveal a novel PTGER2-ß-catenin loop in Treg cells linking environmental high-salt conditions to autoimmunity.


Subject(s)
Autoimmunity/immunology , Inflammation/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , T-Lymphocytes, Regulatory/immunology , beta Catenin/immunology , Animals , Gene Expression Regulation/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Mice, Inbred C57BL , Receptors, Prostaglandin E, EP2 Subtype/immunology , T-Lymphocytes, Regulatory/metabolism
5.
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
6.
J Leukoc Biol ; 102(3): 881-895, 2017 09.
Article in English | MEDLINE | ID: mdl-28630103

ABSTRACT

Dendritic cells (DCs) are APCs essential in regulating the immune response. PGE2, produced during inflammation, has a pivotal role in the maturation of DCs and, therefore, is vital for the immune response. The large variety of biologic functions governed by PGE2 is mediated by its signaling through 4 distinct E-type prostanoid (EP) receptors. Immunogenic DCs express EP2 and EP4, which mediate the PGE2 signaling. However, the expression and function of EP receptors in human tolerogenic DCs (tol-DCs), which present an inhibitory phenotype, have not yet, to our knowledge, been assessed. To clarify the role of EP receptors in tol-DCs, we examined the expression of different EP receptors and their effect using selective agonists in human cells. We find that EP2 and EP3 expression are up-regulated in in vitro-generated tol-DCs compared with mature DCs (mDCs). Activation of EP2-EP4 has a direct effect on the surface expression of costimulatory molecules and maturation receptors, such as CD80, CD83, and CD86 or MHCII and CCR7 in tol-DCs, the latter being exclusively modulated by PGE2-EP4 signaling. Importantly, we find that EP2 and EP3 receptors are involved in tolerance induction through IL-10 production by tol-DCs. These results are in sharp contrast with the inflammatory role of EP4 Moreover, we show that DCs generated in the presence of agonists for EP receptors, induce naive T cell differentiation toward polarized Th1/Th17 cells. Given the differential effects of EP receptors, our results suggest that EP receptor agonist/antagonists might become relevant novel drug templates to modulate immune response.


Subject(s)
Dendritic Cells/immunology , Dinoprostone/pharmacology , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP3 Subtype/immunology , Antigens, CD/immunology , Histocompatibility Antigens Class II/immunology , Humans , Interleukin-10/immunology , Receptors, CCR7/immunology , Th1 Cells/immunology , Th17 Cells/immunology
7.
J Immunol ; 198(8): 3255-3263, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28258202

ABSTRACT

The endocannabinoids 2-arachidonoyl-glycerol and N-arachidonoyl-ethanolamine mediate an array of pro- and anti-inflammatory effects. These effects are related, in part, to their metabolism by eicosanoid biosynthetic enzymes. For example, N-arachidonoyl-ethanolamine and 2-arachidonoyl-glycerol can be metabolized by cyclooxygenase-2 into PG-ethanolamide (PG-EA) and PG-glycerol (PG-G), respectively. Although PGE2 is a recognized suppressor of neutrophil functions, the impact of cyclooxygenase-derived endocannabinoids such as PGE2-EA or PGE2-G on neutrophils is unknown. This study's aim was to define the effects of these mediators on neutrophil functions and the underlying cellular mechanisms involved. We show that PGE2-G, but not PGE2-EA, inhibits leukotriene B4 biosynthesis, superoxide production, migration, and antimicrobial peptide release. The effects of PGE2-G were prevented by EP1/EP2 receptor antagonist AH-6809 but not the EP4 antagonist ONO-AE2-227. The effects of PGE2-G required its hydrolysis into PGE2, were not observed with the non-hydrolyzable PGE2-serinol amide, and were completely prevented by methyl-arachidonoyl-fluorophosphate and palmostatin B, and partially prevented by JZL184 and WWL113. Although we could detect six of the documented PG-G hydrolases in neutrophils by quantitative PCR, only ABHD12 and ABHD16A were detected by immunoblot. Our pharmacological data, combined with our protein expression data, did not allow us to pinpoint one PGE2-G lipase, and rather support the involvement of an uncharacterized lipase and/or of multiple hydrolases. In conclusion, we show that PGE2-G inhibits human neutrophil functions through its hydrolysis into PGE2, and by activating the EP2 receptor. This also indicates that neutrophils could regulate inflammation by altering the balance between PG-G and PG levels in vivo.


Subject(s)
Dinoprostone/metabolism , Endocannabinoids/metabolism , Neutrophils/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Chromatography, Liquid , Dinoprostone/immunology , Endocannabinoids/immunology , Glycerol , Humans , Immunoblotting , Mass Spectrometry , Neutrophils/immunology , Polymerase Chain Reaction , Receptors, Prostaglandin E, EP2 Subtype/immunology
8.
Biochem Biophys Res Commun ; 475(1): 19-24, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27157139

ABSTRACT

Dimethyl fumarate (DMF) was recently approved by the FDA for the treatment of relapsing remitting MS. The pathology of MS is a result of both immune dysregulation and oxidative stress induced damage, and DMF is believed to have therapeutic effects on both of these processes. However, the mechanisms of action of DMF are not fully understood. To determine if DMF is able to activate signaling cascades that affect immune dysregulation, we treated human peripheral blood mononuclear cells with DMF. We discovered that DMF stimulates cyclic adenosine monophosphate (cAMP) production after 1 min treatment in vitro. cAMP is a small molecule second messenger that has been shown to modulate immune response. Using pharmacological inhibitors, we determined that adenylyl cyclase mediates DMF induced cAMP production; DMF activated the prostaglandin EP2 receptor to produce cAMP. This response was not due to increased endogenous production of prostaglandin E2 (PGE2), but was enhanced by addition of exogenous PGE2. Furthermore, we determined that the bioactive metabolite of DMF, monomethyl fumarate (MMF), also stimulates cAMP production. These novel findings suggest that DMF may provide protection against MS by inhibiting immune cell function via the cAMP signaling pathway.


Subject(s)
Cyclic AMP/immunology , Dimethyl Fumarate/pharmacology , Immunosuppressive Agents/pharmacology , Leukocytes, Mononuclear/drug effects , Receptors, Prostaglandin E, EP2 Subtype/immunology , Signal Transduction/drug effects , Adenylyl Cyclases/immunology , Dinoprostone/immunology , Humans , Leukocytes, Mononuclear/immunology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology
9.
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
10.
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
11.
PLoS Negl Trop Dis ; 9(8): e0004025, 2015.
Article in English | MEDLINE | ID: mdl-26305786

ABSTRACT

Inflammation plays an important role in the pathophysiology of Chagas disease, caused by Trypanosoma cruzi. Prostanoids are regulators of homeostasis and inflammation and are produced mainly by myeloid cells, being cyclooxygenases, COX-1 and COX-2, the key enzymes in their biosynthesis from arachidonic acid (AA). Here, we have investigated the expression of enzymes involved in AA metabolism during T. cruzi infection. Our results show an increase in the expression of several of these enzymes in acute T. cruzi infected heart. Interestingly, COX-2 was expressed by CD68+ myeloid heart-infiltrating cells. In addition, infiltrating myeloid CD11b+Ly6G- cells purified from infected heart tissue express COX-2 and produce prostaglandin E2 (PGE2) ex vivo. T. cruzi infections in COX-2 or PGE2-dependent prostaglandin receptor EP-2 deficient mice indicate that both, COX-2 and EP-2 signaling contribute significantly to the heart leukocyte infiltration and to the release of chemokines and inflammatory cytokines in the heart of T. cruzi infected mice. In conclusion, COX-2 plays a detrimental role in acute Chagas disease myocarditis and points to COX-2 as a potential target for immune intervention.


Subject(s)
Chagas Disease/immunology , Cyclooxygenase 2/immunology , Dinoprostone/immunology , Myocarditis/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Trypanosoma cruzi/physiology , Animals , Chagas Disease/complications , Chagas Disease/enzymology , Chagas Disease/genetics , Cyclooxygenase 2/genetics , Cytokines/genetics , Cytokines/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/etiology , Myocarditis/genetics , Myocardium/enzymology , Myocardium/immunology , Receptors, Prostaglandin E, EP2 Subtype/genetics , Trypanosoma cruzi/immunology
12.
Int Immunopharmacol ; 28(1): 376-81, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26122137

ABSTRACT

The present study aimed to explore whether the inhibition of prostaglandin E2 enhances pulmonary anti-Cryptococcus neoformans immunity. Lung colony forming unit (CFU) assays demonstrated that the cryptococcal infection was dramatically depressed in mice given EP2 and EP4 or single EP antagonist treatment compared to the untreated wild type mice (p<0.05), leading to the increased survival of the infected mice by 8-9 days or 2-4 days, respectively. RT-PCR and flow cytometry assays showed that the expression of IFN-γ, IL-17, IL-22 in M1 macrophages and IL-10 in M2 macrophages increased significantly at 1 week post-infection in mice with either EP2 or EP4 blockade (p<0.05). The polarization of alveolar macrophages showed that, at 1 week post infection, the alveolar macrophages in untreated wild type mice, TLR4(-/-) mice and TLR4(-/-) mice with EP2 and EP4 blockade were strongly M2 polarized, whereas the alveolar macrophages in wild type mice with EP2 and EP4 blockade were M1 polarized. In conclusion, the blockade of EP2 and EP4 promotes mouse survival after cryptococcus infection by promoting the production of cytokines via TLR4, as well as the enhanced M1 polarization of alveolar macrophages.


Subject(s)
Cryptococcosis/immunology , Cryptococcus neoformans , Dinoprostone/antagonists & inhibitors , Macrophages, Alveolar/immunology , Toll-Like Receptor 4/immunology , Animals , Cryptococcosis/microbiology , Cytokines/genetics , Cytokines/immunology , Dinoprostone/immunology , Lung/immunology , Lung/microbiology , Mice, Inbred BALB C , Mice, Knockout , Prostaglandin Antagonists/pharmacology , RNA, Messenger/metabolism , 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 , Xanthones/pharmacology
13.
Clin Exp Allergy ; 45(10): 1590-600, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25823713

ABSTRACT

BACKGROUND: Prostaglandin E2 (PGE2 ) has been proposed to exert antiasthmatic effects in patients, to prevent antigen-induced airway pathology in murine models, and to inhibit mast cells (MC) activity in vitro. OBJECTIVE: To assess in a murine model whether the protective effect of PGE2 may be a consequence of its ability to activate the E-prostanoid (EP)2 receptor on airway MC. METHODS: Either BALB/c or C57BL/6 mice were exposed intranasally (i.n.) to house dust mite (HDM) aeroallergens. Both strains were given PGE2 locally (0.3 mg/kg), but only BALB/c mice were administered butaprost (EP2 agonist: 0.3 mg/kg), or AH6809 (EP2 antagonist; 2.5 mg/kg) combined with the MC stabilizer sodium cromoglycate (SCG: 25 mg/kg). Airway hyperresponsiveness (AHR) and inflammation, along with lung MC activity, were evaluated. In addition, butaprost's effect was assessed in MC-mediated passive cutaneous anaphylaxis (PCA) in mice challenged with 2,4-dinitrophenol (DNP). RESULTS: Selective EP2 agonism attenuated aeroallergen-caused AHR and inflammation in HDM-exposed BALB/c mice, and this correlated with a reduced lung MC activity. Accordingly, the blockade of endogenous PGE2 by means of AH6809 worsened airway responsiveness in sensitive BALB/c mice, and such worsening was reversed by SCG. The relevance of MC to PGE2 -EP2 driven protection was further highlighted in MC-dependent PCA, where butaprost fully prevented MC-induced ear swelling. Unlike in BALB/c mice, PGE2 did not protect the airways of HDM-sensitized C57BL/6 animals, a strain in which we showed MC to be irrelevant to aeroallergen-driven AHR and inflammation. CONCLUSIONS & CLINICAL RELEVANCE: The beneficial effect of both exogenous and endogenous PGE2 in aeroallergen-sensitized mice may be attributable to the activation of the EP2 receptor, which in turn acts as a restrainer of airway MC activity. This opens a path towards the identification of therapeutic targets against asthma along the 'EP2 -MC-airway' axis.


Subject(s)
Asthma/immunology , Dinoprostone/immunology , Mast Cells/immunology , Pyroglyphidae , Receptors, Prostaglandin E, EP2 Subtype/immunology , Animals , Asthma/pathology , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/pathology , Mast Cells/pathology , Mice , Mice, Inbred BALB C
14.
Mol Immunol ; 63(1): 61-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24768319

ABSTRACT

Despite the fact that cyclooxygenase and its products, prostaglandins, have been traditionally associated with the development of inflammation, PGE2 was implicated early on as potentially beneficial in asthma. During the 1970s and 1980s, several studies reported the bronchodilator effect of PGE2 in asthma patients. In parallel, it was being shown to exert an inhibitory effect on mast cells in vitro. In spite of this, data supporting the beneficial role for PGE2 in asthma were scarce and sometimes controversial. Many years later, in vitro and in vivo studies suggested a range of biological activities attributable to PGE2, others than the ability to relax smooth muscle, that potentially explained some of the observed positive effects in asthma. The identification and cloning of the four PGE2 receptors made available new tools with which to fine-tune investigation of the anti-inflammatory, pro-inflammatory, immunoregulatory, and bronchodilation mechanisms of PGE2. Among these, several suggested involvement of mast cells, a cell population known to play a fundamental role in acute and chronic asthma. Indeed, it has been shown that PGE2 prevents human and murine MC activity in vitro through activation of the EP2 receptor, and also that both exogenously administered and endogenous PGE2 inhibit airway MC activity in vivo in mouse models of asthma (likely through an EP2-mediated mechanism as well). In the last few years, we have furthered into the functional connection between PGE2-induced mast cells inhibition and attenuated damage, in asthma and allergy models. The validity of the findings supporting a beneficial effect of PGE2 in different asthma phases, the direct effect of PGE2 on mast cells populations, and the functional implications of the PGE2-MC interaction on airway function are some of the topics addressed in this review, under the assumption that increased understanding of the PGE2-EP2-mast cell axis will likely lead to the discovery of novel antiasthma targets.


Subject(s)
Asthma/drug therapy , Asthma/immunology , Dinoprostone/immunology , Dinoprostone/therapeutic use , Mast Cells/drug effects , Mast Cells/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Bronchodilator Agents/therapeutic use , Disease Models, Animal , Humans , Inflammation/immunology , Mice , Respiratory System/immunology
15.
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
16.
Bull Math Biol ; 76(6): 1352-75, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24809944

ABSTRACT

Cyclic AMP is important for the resolution of inflammation, as it promotes anti-inflammatory signaling in several immune cell lines. In this paper, we present an immune cell specific model of the cAMP signaling cascade, paying close attention to the specific isoforms of adenylyl cyclase (AC) and phosphodiesterase that control cAMP production and degradation, respectively, in these cells. The model describes the role that G protein subunits, including Gαs, Gαi, and Gßγ, have in regulating cAMP production. Previously, Gαi activation has been shown to increase the level of cAMP in certain immune cell types. This increase in cAMP is thought to be mediated by ßγ subunits which are released upon Gα activation and can directly stimulate specific isoforms of AC. We conduct numerical experiments in order to explore the mechanisms through which Gαi activation can increase cAMP production. An important conclusion of our analysis is that the relative abundance of different G protein subunits is an essential determinant of the cAMP profile in immune cells. In particular, our model predicts that limited availability of ßγ subunits may both (i) enable immune cells to link inflammatory Gαi signaling to anti-inflammatory cAMP production thereby creating a balanced immune response to stimulation with low concentrations of PGE2, and (ii) prohibit robust anti-inflammatory cAMP signaling in response to stimulation with high concentrations of PGE2.


Subject(s)
Adenylyl Cyclases/immunology , Cyclic AMP/immunology , Models, Immunological , Signal Transduction/immunology , Cell Line , Computer Simulation , GTP-Binding Protein alpha Subunits/immunology , GTP-Binding Protein beta Subunits/immunology , GTP-Binding Protein gamma Subunits/immunology , Kinetics , Receptor, Anaphylatoxin C5a/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology
17.
J Allergy Clin Immunol ; 133(2): 379-87, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24075232

ABSTRACT

BACKGROUND: Endogenous prostanoids have been suggested to modulate sensitization during experimental allergic asthma, but the specific role of prostaglandin (PG) E2 or of specific E prostanoid (EP) receptors is not known. OBJECTIVE: Here we tested the role of EP2 signaling in allergic asthma. METHODS: Wild-type (WT) and EP2(-/-) mice were subjected to ovalbumin sensitization and acute airway challenge. The PGE2 analog misoprostol was administered during sensitization in both genotypes. In vitro culture of splenocytes and flow-sorted dendritic cells and T cells defined the mechanism by which EP2 exerted its protective effect. Adoptive transfer of WT and EP2(-/-) CD4 T cells was used to validate the importance of EP2 expression on T cells. RESULTS: Compared with WT mice, EP2(-/-) mice had exaggerated airway inflammation in this model. Splenocytes and lung lymph node cells from sensitized EP2(-/-) mice produced more IL-13 than did WT cells, suggesting increased sensitization. In WT but not EP2(-/-) mice, subcutaneous administration of misoprostol during sensitization inhibited allergic inflammation. PGE2 decreased cytokine production and inhibited signal transducer and activator of transcription 6 phosphorylation by CD3/CD28-stimulated CD4(+) T cells. Coculture of flow cytometry-sorted splenic CD4(+) T cells and CD11c(+) dendritic cells from WT or EP2(-/-) mice suggested that the increased IL-13 production in EP2(-/-) mice was due to the lack of EP2 specifically on T cells. Adoptive transfer of CD4(+) EP2(-/-) T cells caused greater cytokine production in the lungs of WT mice than did transfer of WT CD4(+) T cells. CONCLUSION: We conclude that the PGE2-EP2 axis is an important endogenous brake on allergic airway inflammation and primarily targets T cells and that its agonism represents a potential novel therapeutic approach to asthma.


Subject(s)
Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , Dinoprostone/immunology , Pneumonia/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Adoptive Transfer , Allergens , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Cytokines/immunology , Dendritic Cells/immunology , Lymph Nodes/immunology , Male , Mice , Mice, Knockout , Misoprostol/pharmacology , Ovalbumin , Receptors, Prostaglandin E, EP2 Subtype/genetics , Spleen/immunology
18.
Mol Immunol ; 54(3-4): 284-95, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23337716

ABSTRACT

Dendritic cells (DCs) are central players in coordinating immune responses, both innate and adaptive. While the role of lipid mediators in the immune response has been the subject of many investigations, the precise role of prostaglandins has often been plagued by contradictory studies. In this study, we examined the role of PGE(2) on human DC function. Although studies have suggested that PGE(2) specifically plays a role in DC motility and cytokine release profile, the precise receptor usage and signaling pathways involved remain unclear. In this report we found that irrespective of the human donor, monocyte-derived dendritic cells (MoDCs) express three of the four PGE(2) receptor subtypes (EP(2-4)), although only EP(2) and EP(4) were active with respect to cytokine production. Using selective EP receptor antagonists and agonists, we demonstrate that PGE(2) coordinates control of IL-23 release (a promoter of Th17, an autoimmune associated T cell subset) in a dose-dependent manner by differential use of EP(2) and EP(4) receptors in LPS-activated MoDCs. This is in contrast to IL-12, which is dose dependently inhibited by PGE(2) through both receptor subtypes. Low concentrations (∼1-10nM) of PGE(2) promoted IL-23 production via EP(4) receptors, while at higher (>50 nM), but still physiologically relevant concentrations, IL-23 is suppressed by an EP(2) dependent mechanism. These results can be explained by differential regulation of the common subunit, IL-12p40, and IL-23p19, by EP(2) and EP(4). By these means, PGE(2) can act as a regulatory switch of immune responses depending on its concentration in the microenvironment. In addition, we believe these results may also explain why seemingly conflicting biological functions assigned to PGE(2) have been reported in the literature, as the concentration of ligand (PGE(2)) fundamentally alters the nature of the response. This finding also highlights the potential of designing therapeutics which differentially target these receptors.


Subject(s)
Dendritic Cells/immunology , Dinoprostone/immunology , Monocytes/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Cyclic AMP/immunology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dinoprostone/metabolism , Guanine Nucleotide Exchange Factors/immunology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Interleukin-23 Subunit p19/immunology , Interleukin-23 Subunit p19/metabolism , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Prostaglandin E, EP2 Subtype/biosynthesis , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/biosynthesis , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism
20.
J Infect Dis ; 206(12): 1816-25, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23033144

ABSTRACT

Mycobacterium tuberculosis, the causative agent of tuberculosis, resides and replicates within susceptible hosts by inhibiting host antimicrobial mechanisms. Prostaglandin E(2) (PGE(2)), produced by M. tuberculosis-infected macrophages, exerts a variety of immunomodulatory functions via 4 receptors (EP1-EP4), each mediating distinct PGE(2) functions. Here, we show that M. tuberculosis infection selectively upregulates EP2 messenger RNA expression in CD4(+) T cells. We found that EP2 deficiency in mice increases susceptibility to M. tuberculosis infection, which correlated with reduced antigen-specific T-cell responses and increased levels of CD4(+)CD25(+)Foxp3(+) T-regulatory cells. These findings have revealed an important role for EP2 in host immune defense against tuberculosis. As a G protein-coupled receptor, EP2 could serve as a target for immunotherapy of tuberculosis.


Subject(s)
Mycobacterium tuberculosis/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Tuberculosis/genetics , Tuberculosis/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Forkhead Transcription Factors/analysis , Gene Expression , Gene Expression Profiling , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/analysis , Mice , Mice, Inbred C57BL , Receptors, Prostaglandin E, EP2 Subtype/biosynthesis , Receptors, Prostaglandin E, EP2 Subtype/genetics , T-Lymphocytes, Regulatory/chemistry , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...