Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Pharmacol Rev ; 72(4): 910-968, 2020 10.
Article in English | MEDLINE | ID: mdl-32962984

ABSTRACT

Prostaglandins are derived from arachidonic acid metabolism through cyclooxygenase activities. Among prostaglandins (PGs), prostacyclin (PGI2) and PGE2 are strongly involved in the regulation of homeostasis and main physiologic functions. In addition, the synthesis of these two prostaglandins is significantly increased during inflammation. PGI2 and PGE2 exert their biologic actions by binding to their respective receptors, namely prostacyclin receptor (IP) and prostaglandin E2 receptor (EP) 1-4, which belong to the family of G-protein-coupled receptors. IP and EP1-4 receptors are widely distributed in the body and thus play various physiologic and pathophysiologic roles. In this review, we discuss the recent advances in studies using pharmacological approaches, genetically modified animals, and genome-wide association studies regarding the roles of IP and EP1-4 receptors in the immune, cardiovascular, nervous, gastrointestinal, respiratory, genitourinary, and musculoskeletal systems. In particular, we highlight similarities and differences between human and rodents in terms of the specific roles of IP and EP1-4 receptors and their downstream signaling pathways, functions, and activities for each biologic system. We also highlight the potential novel therapeutic benefit of targeting IP and EP1-4 receptors in several diseases based on the scientific advances, animal models, and human studies. SIGNIFICANCE STATEMENT: In this review, we present an update of the pathophysiologic role of the prostacyclin receptor, prostaglandin E2 receptor (EP) 1, EP2, EP3, and EP4 receptors when activated by the two main prostaglandins, namely prostacyclin and prostaglandin E2, produced during inflammatory conditions in human and rodents. In addition, this comparison of the published results in each tissue and/or pathology should facilitate the choice of the most appropriate model for the future studies.


Subject(s)
Receptors, Prostaglandin E/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Dinoprostone/immunology , Dinoprostone/metabolism , Epoprostenol/immunology , Epoprostenol/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Mice , Polymorphism, Single Nucleotide , Protein Multimerization , Rats , Receptors, Prostaglandin E/chemistry , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/immunology , Species Specificity
2.
J Allergy Clin Immunol ; 141(5): 1761-1773.e6, 2018 05.
Article in English | MEDLINE | ID: mdl-29217133

ABSTRACT

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) are involved in the initial phase of type 2 inflammation and can amplify allergic immune responses by orchestrating other type 2 immune cells. Prostaglandin (PG) E2 is a bioactive lipid that plays protective roles in the lung, particularly during allergic inflammation. OBJECTIVE: We set out to investigate how PGE2 regulates human ILC2 function. METHODS: The effects of PGE2 on human ILC2 proliferation and intracellular cytokine and transcription factor expression were assessed by means of flow cytometry. Cytokine production was measured by using ELISA, and real-time quantitative PCR was performed to detect PGE2 receptor expression. RESULTS: PGE2 inhibited GATA-3 expression, as well as production of the type 2 cytokines IL-5 and IL-13, from human tonsillar and blood ILC2s in response to stimulation with a combination of IL-25, IL-33, thymic stromal lymphopoietin, and IL-2. Furthermore, PGE2 downregulated the expression of IL-2 receptor α (CD25). In line with this observation, PGE2 decreased ILC2 proliferation. These effects were mediated by the combined action of E-type prostanoid receptor (EP) 2 and EP4 receptors, which were specifically expressed on ILC2s. CONCLUSION: Our findings reveal that PGE2 limits ILC2 activation and propose that selective EP2 and EP4 receptor agonists might serve as a promising therapeutic approach in treating allergic diseases by suppressing ILC2 function.


Subject(s)
Dinoprostone/immunology , Gene Expression Regulation/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Cells, Cultured , Cytokines/immunology , GATA3 Transcription Factor/immunology , Humans , Inflammation/immunology , Receptors, Prostaglandin E/immunology
3.
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
4.
Nucleic Acids Res ; 44(6): 2888-97, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26825461

ABSTRACT

Fever is commonly used to diagnose disease and is consistently associated with increased mortality in critically ill patients. However, the molecular controls of elevated body temperature are poorly understood. We discovered that the expression of RNA-binding motif protein 3 (RBM3), known to respond to cold stress and to modulate microRNA (miRNA) expression, was reduced in 30 patients with fever, and in THP-1-derived macrophages maintained at a fever-like temperature (40 °C). Notably, RBM3 expression is reduced during fever whether or not infection is demonstrable. Reduced RBM3 expression resulted in increased expression of RBM3-targeted temperature-sensitive miRNAs, we termed thermomiRs. ThermomiRs such as miR-142-5p and miR-143 in turn target endogenous pyrogens including IL-6, IL6ST, TLR2, PGE2 and TNF to complete a negative feedback mechanism, which may be crucial to prevent pathological hyperthermia. Using normal PBMCs that were exogenously exposed to fever-like temperature (40 °C), we further demonstrate the trend by which decreased levels of RBM3 were associated with increased levels of miR-142-5p and miR-143 and vice versa over a 24 h time course. Collectively, our results indicate the existence of a negative feedback loop that regulates fever via reduced RBM3 levels and increased expression of miR-142-5p and miR-143.


Subject(s)
Feedback, Physiological , Fever/genetics , Leukocytes, Mononuclear/immunology , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Body Temperature , Body Temperature Regulation/genetics , Cell Line , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/immunology , Fever/immunology , Fever/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Macrophages/cytology , Macrophages/immunology , MicroRNAs/immunology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/immunology , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/immunology , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
5.
Biochim Biophys Acta ; 1851(4): 414-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25038274

ABSTRACT

Prostaglandin E2 (PGE2) is one of the most typical lipid mediators produced from arachidonic acid (AA) by cyclooxygenase (COX) as the rate-limiting enzyme, and acts on four kinds of receptor subtypes (EP1-EP4) to elicit its diverse actions including pyrexia, pain sensation, and inflammation. Recently, the molecular mechanisms underlying the PGE2 actions mediated by each EP subtype have been elucidated by studies using mice deficient in each EP subtype as well as several compounds highly selective to each EP subtype, and their findings now enable us to discuss how PGE2 initiates and exacerbates inflammation at the molecular level. Here, we review the recent advances in PGE2 receptor research by focusing on the activation of mast cells via the EP3 receptor and the control of helper T cells via the EP2/4 receptor, which are the molecular mechanisms involved in PGE2-induced inflammation that had been unknown for many years. We also discuss the roles of PGE2 in acute inflammation and inflammatory disorders, and the usefulness of anti-inflammatory therapies that target EP receptors. This article is part of a Special Issue entitled "Oxygenated metabolism of PUFA: analysis and biological relevance".


Subject(s)
Dinoprostone/metabolism , Inflammation/metabolism , Receptors, Prostaglandin E/metabolism , Signal Transduction , Animals , Anti-Inflammatory Agents/pharmacology , Dinoprostone/chemistry , Dinoprostone/immunology , Drug Design , Humans , Inflammation/immunology , Inflammation/prevention & control , Lymphocyte Activation , Mast Cells/immunology , Mast Cells/metabolism , Molecular Structure , Molecular Targeted Therapy , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Structure-Activity Relationship , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
6.
Int Immunopharmacol ; 23(1): 163-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25196430

ABSTRACT

Prostaglandin E2 (PGE2), a very potent lipid mediator produced from arachidonic acid (AA) through the action of cyclooxygenase (COX) enzymes, is implicated in the regulation of dendritic cell (DC) functions such as differentiation ability, cytokine-producing capacity, Th-cell polarizing ability, migration and maturation. DCs are the most potent antigen-presenting cells and play major roles in both the induction of primary immune responses and tolerance. It is well established that PGE2 functions significantly in the pathogenesis of rheumatoid arthritis (RA). Although the role of PGE2 in RA has been studied extensively, the effects of PGE2 on DC biology and the role of DCs in RA have not become the focus of investigation until recently. Here, we summarize the latest progress in PGE2 research with respect to DC functions, as well as the role of PGE2 receptor signaling of DCs in the pathogenesis of RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Dendritic Cells/immunology , Receptors, Prostaglandin E/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Antigen Presentation , Cell Differentiation , Cytokines/metabolism , Humans , Immune Tolerance , Immunity, Cellular , Lipid Metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Signal Transduction
7.
Parasite Immunol ; 35(1): 51-4, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23075034

ABSTRACT

Through their receptors, prostaglandins play crucial roles in various infections. Although prostaglandin E2 (PGE2) is implicated as a susceptibility factor in Leishmania infection, the relative contributions of its four receptors--EP1, EP2, EP3 and EP4--to this infection remain unknown. We report that Leishmania major infection of BALB/c-derived peritoneal macrophages up-regulated EP1 and EP3 expressions but down-regulated EP2 and EP4 expressions. EP2 and EP4 agonists reduced parasite load, but EP1 and EP3 agonists increased parasite load in macrophages in vitro. Agonists of EP2 and EP4, antagonists of EP1 and EP3, or lentivirally expressed EP1-shRNA and EP3-shRNA significantly reduced parasite burden in susceptible BALB/c mice. These novel data suggest differential regulation and counteractive functions of EP receptor subsets.


Subject(s)
Leishmania major/physiology , Leishmaniasis, Cutaneous/immunology , Macrophages, Peritoneal/immunology , Receptors, Prostaglandin E/immunology , Animals , Leishmaniasis, Cutaneous/parasitology , Mice , Mice, Inbred BALB C , Parasite Load , RNA, Small Interfering , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/classification
8.
Proc Natl Acad Sci U S A ; 107(27): 12233-8, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20566843

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis (MS). Although prostaglandin (PG) concentrations are increased in cerebrospinal fluid of MS patients, the role of PGs in MS is unknown. We examined this issue by subjecting mice deficient in each PG receptor type or subtype to EAE induction and using agonists or antagonists selective for each of the four PGE receptor (EP) subtypes. Among PG receptor-deficient mice, only EP4(-/-) mice manifested significant suppression of EAE, which was mimicked in wild-type mice and to a greater extent, in EP2(-/-) mice by administration of the EP4 antagonist ONO-AE3-208 during the immunization phase. EP4 antagonism during immunization also suppressed the generation of antigen-specific T helper (Th) 1 and Th17 cells in wild-type mice and to a greater extent, in EP2(-/-) mice. ONO-AE3-208 administration at EAE onset had little effect on disease severity, and its administration throughout the experimental period did not cause significant reduction of the peak of disease, suggesting that, in addition to its facilitative action during the immunization phase, EP4 exerts a preventive action in the elicitation phase. Administration of the EP4 agonist ONO-AE1-329 at EAE onset delayed and suppressed disease progression as well as inhibited the associated increase in permeability of the blood-brain barrier. Thus, PGE(2) exerts dual functions in EAE, facilitating Th1 and Th17 cell generation redundantly through EP4 and EP2 during immunization and attenuating invasion of these cells into the brain by protecting the blood-brain barrier through EP4.


Subject(s)
Dinoprostone/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Receptors, Prostaglandin E/immunology , Signal Transduction/immunology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Humans , Interferon-gamma/metabolism , Interleukin-17/metabolism , Male , Methyl Ethers/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Naphthalenes/pharmacology , Phenylbutyrates/pharmacology , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Signal Transduction/drug effects , Signal Transduction/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism
9.
J Immunol ; 184(12): 7207-18, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20483760

ABSTRACT

Peripheral inflammation leads to immune responses in brain characterized by microglial activation, elaboration of proinflammatory cytokines and reactive oxygen species, and secondary neuronal injury. The inducible cyclooxygenase (COX), COX-2, mediates a significant component of this response in brain via downstream proinflammatory PG signaling. In this study, we investigated the function of the PGE2 E-prostanoid (EP) 4 receptor in the CNS innate immune response to the bacterial endotoxin LPS. We report that PGE2 EP4 signaling mediates an anti-inflammatory effect in brain by blocking LPS-induced proinflammatory gene expression in mice. This was associated in cultured murine microglial cells with decreased Akt and I-kappaB kinase phosphorylation and decreased nuclear translocation of p65 and p50 NF-kappaB subunits. In vivo, conditional deletion of EP4 in macrophages and microglia increased lipid peroxidation and proinflammatory gene expression in brain and in isolated adult microglia following peripheral LPS administration. Conversely, EP4 selective agonist decreased LPS-induced proinflammatory gene expression in hippocampus and in isolated adult microglia. In plasma, EP4 agonist significantly reduced levels of proinflammatory cytokines and chemokines, indicating that peripheral EP4 activation protects the brain from systemic inflammation. The innate immune response is an important component of disease progression in a number of neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In addition, recent studies demonstrated adverse vascular effects with chronic administration of COX-2 inhibitors, indicating that specific PG signaling pathways may be protective in vascular function. This study supports an analogous and beneficial effect of PGE2 EP4 receptor signaling in suppressing brain inflammation.


Subject(s)
Brain/immunology , Immunity, Innate/immunology , Inflammation Mediators/immunology , Receptors, Prostaglandin E/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Brain/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression , Gene Expression Regulation/immunology , Immunohistochemistry , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/metabolism , Microscopy, Confocal , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP4 Subtype , Reverse Transcriptase Polymerase Chain Reaction
10.
Int Immunopharmacol ; 9(5): 534-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19539567

ABSTRACT

Limited data are available on the mechanisms that constrain the function of regulatory populations of T cells. Prostaglandin E2 (PGE2) is an endogenous membrane phospholipid metabolite that has important immunomodulatory effects on T cell function. Our previous microarray data indicated that E prostanoid receptor 2 (EP2), a receptor for PGE2, is expressed by regulatory alphabetaTCR(+) CD4(-) CD8(-) NK1.1(-) double negative T (DN Treg) cell clones but not by their non-regulatory natural mutants. Hence, the hypothesis that PGE2 may influence DN Treg cell proliferation and/or regulatory function was tested in this study. Our data indicate that PGE2 acts via the EP2 receptor on DN Treg cells to inhibit their proliferation, an effect reproduced by the EP2-specific agonist butaprost and abrogated by the EP2 antagonist AH6809. In contrast, PGE2 did not affect the ability of DN Treg cells to kill syngeneic CD8(+) T cells activated by allogeneic stimulation. Together, these findings suggest a role for PGE2 in limiting the expansion of DN Treg cells.


Subject(s)
Dinoprostone/metabolism , Receptors, Prostaglandin E/metabolism , T-Lymphocytes, Regulatory/metabolism , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , CD4 Antigens , CD8 Antigens , Cell Proliferation/drug effects , Clone Cells , Cytotoxicity, Immunologic/drug effects , Dinoprostone/immunology , Immunomagnetic Separation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostaglandin Antagonists/pharmacology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E, EP2 Subtype , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Xanthones/pharmacology
11.
Nat Med ; 15(6): 633-40, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19465928

ABSTRACT

Two distinct helper T (TH) subsets, TH1 and TH17, mediate tissue damage and inflammation in animal models of various immune diseases such as multiple sclerosis, rheumatoid arthritis, inflammatory bowel diseases and allergic skin disorders. These experimental findings, and the implication of these TH subsets in human diseases, suggest the need for pharmacological measures to manipulate these TH subsets. Here we show that prostaglandin E2 (PGE2) acting on its receptor EP4 on T cells and dendritic cells not only facilitates TH1 cell differentiation but also amplifies interleukin-23-mediated TH17 cell expansion in vitro. Administration of an EP4-selective antagonist in vivo decreases accumulation of both TH1 and TH17 cells in regional lymph nodes and suppresses the disease progression in mice subjected to experimental autoimmune encephalomyelitis or contact hypersensitivity. Thus, PGE2-EP4 signaling promotes immune inflammation through TH1 differentiation and TH17 expansion, and EP4 antagonism may be therapeutically useful for various immune diseases.


Subject(s)
Cell Differentiation/immunology , Dinoprostone/immunology , Interleukin-17/immunology , Receptors, Prostaglandin E/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis, Contact/immunology , Dinoprostone/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Inflammation/metabolism , Interleukin-23/biosynthesis , Interleukin-23/immunology , Mice , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP4 Subtype , Th1 Cells/metabolism
12.
Eur J Immunol ; 39(5): 1301-12, 2009 May.
Article in English | MEDLINE | ID: mdl-19384872

ABSTRACT

The contribution of Th1 and Th17 cells in chronic inflammatory conditions leading to autoimmunity remains highly controversial. In inflamed tissues, production of prostaglandins by COX-2 has been proposed to favor Th17 responses indirectly by increasing IL-23 and blocking IL-12 release from APC. We report here that prostaglandin E2 (PGE2) can directly modulate cytokine production by human memory CD4(+) T cells. TCR triggering in the presence of PGE2 increased IL-17 and reduced IFN-gamma production by freshly isolated memory T cells or T-cell clones. PGE2 triggered the EP2 and EP4 receptors expressed on T cells leading to a rapid increase of retinoic-acid-related orphan receptor-gammat (ROR-gammat) and decrease of T-cell-specific T-box transcription factor 21 (T-bet) mRNA. Moreover, PGE2 promoted the selective enrichment of IL-17-producing cells by differentially modulating the proliferation of memory T-cell subsets in vitro. Taken together our results indicate that T-cell effector function is a direct target for PGE2 modulation and suggest a novel mechanism by which inhibitors of prostaglandin synthesis, such as COX-2 inhibitors, exert their anti-inflammatory effect.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Dinoprostone/pharmacology , Interferon-gamma/immunology , Interleukin-17/immunology , T-Box Domain Proteins/immunology , Autoimmunity/drug effects , Autoimmunity/immunology , Dinoprostone/immunology , Humans , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-17/antagonists & inhibitors , Interleukin-17/biosynthesis , Interleukin-17/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/immunology , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/genetics , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Th1 Cells/drug effects , Th1 Cells/immunology
13.
Eur J Immunol ; 39(3): 752-62, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19197941

ABSTRACT

The physiological functions of human TCRVgamma9Vdelta2(+) gammadelta lymphocytes reactive to non-peptide phosphoantigens contribute to cancer immunosurveillance and immunotherapy. However, their regulation by mesenchymal stem cells (MSC), multipotent and immunomodulatory progenitor cells able to infiltrate tumors, has not been investigated so far. By analyzing freshly isolated TCRVgamma9Vdelta2(+) lymphocytes and primary cell lines stimulated with synthetic phosphoantigen or B-cell lymphoma cell lines in the presence of MSC, we demonstrated that MSC were potent suppressors of gammadelta-cell proliferation, cytokine production and cytolytic responses in vitro. This inhibition was mediated by the COX-2-dependent production of prostaglandin E2 (PGE(2)) and by MSC through EP2 and EP4 inhibitory receptors expressed by Vgamma9Vdelta2 T lymphocytes. COX-2 expression and PGE(2) production by MSC were not constitutive, but were induced by IFN-gamma and TNF-alpha secreted by activated Vgamma9Vdelta2 T cells. This regulatory cross-talk between MSC and Vgamma9Vdelta2 T lymphocytes involving PGE(2) could be of importance for the antitumor and antimicrobial activities of gammadelta T cells.


Subject(s)
Cell Communication/immunology , Cyclooxygenase 2/immunology , Mesenchymal Stem Cells/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Dinoprostone/pharmacology , Humans , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/metabolism , Receptor Cross-Talk/immunology , Receptors, Antigen, T-Cell, gamma-delta/antagonists & inhibitors , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/pharmacology
14.
J Immunol ; 181(10): 7273-83, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981149

ABSTRACT

The accumulation of eosinophils in lung tissue is a hallmark of asthma, and it is believed that eosinophils play a crucial pathogenic role in allergic inflammation. Prostaglandin (PG) E(2) exerts anti-inflammatory and bronchoprotective mechanisms in asthma, but the underlying mechanisms have remained unclear. In this study we show that PGE(2) potently inhibits the chemotaxis of purified human eosinophils toward eotaxin, PGD(2), and C5a. Activated monocytes similarly attenuated eosinophil migration, and this was reversed after pretreatment of the monocytes with a cyclooxygenase inhibitor. The selective E-prostanoid (EP) 2 receptor agonist butaprost mimicked the inhibitory effect of PGE(2) on eosinophil migration, whereas an EP2 antagonist completely prevented this effect. Butaprost, and also PGE(2), inhibited the C5a-induced degranulation of eosinophils. Moreover, selective kinase inhibitors revealed that the inhibitory effect of PGE(2) on eosinophil migration depended upon activation of PI3K and protein kinase C, but not cAMP. In animal models, the EP2 agonist butaprost inhibited the rapid mobilization of eosinophils from bone marrow of the in situ perfused guinea pig hind limb and prevented the allergen-induced bronchial accumulation of eosinophils in OVA-sensitized mice. Immunostaining showed that human eosinophils express EP2 receptors and that EP2 receptor expression in the murine lungs is prominent in airway epithelium and, after allergen challenge, in peribronchial infiltrating leukocytes. In summary, these data show that EP2 receptor agonists potently inhibit eosinophil trafficking and activation and might hence be a useful therapeutic option in eosinophilic diseases.


Subject(s)
Chemotaxis, Leukocyte/immunology , Dinoprostone/immunology , Eosinophils/immunology , Receptors, Prostaglandin E/immunology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Eosinophils/drug effects , Eosinophils/metabolism , Guinea Pigs , Humans , Immunohistochemistry , Mice , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/immunology , Protein Kinase C/metabolism , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP2 Subtype
15.
J Immunol ; 181(8): 5637-45, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832722

ABSTRACT

Monocytes/macrophages committed to death by peroxynitrite nevertheless survive with a signaling response promoting Bad phosphorylation, as well as its cytosolic localization, via upstream activation of cytosolic phospholipase A(2), 5-lipoxygenase, and protein kinase C alpha. We now report evidence for an alternative mechanism converging in Bad phosphorylation when the expression/activity of the above enzymes are suppressed. Under these conditions, also associated with peroxynitrite-dependent severe inhibition of Akt, an additional Bad kinase, Bad dephosphorylation promoted its accumulation in the mitochondria and a prompt lethal response. PGE(2) prevented toxicity via EP(2) receptor-mediated protein kinase A-dependent Bad phosphorylation. This notion was established in U937 cells by the following criteria: 1) there was a strong correlation between survival and cAMP accumulation, both in the absence and presence of phosphodiesterase inhibitors; 2) direct activation of adenylyl cyclase afforded cytoprotection; and 3) PGE(2) promoted loss of mitochondrial Bad and cytoprotection, mimicked by EP(2) receptor agonists, and prevented by EP(2) receptor antagonists or protein kinase A inhibitors. Finally, selected experiments performed in human monocytes/macrophages and in rat peritoneal macrophages indicated that the above cytoprotective pathway is a general response of cells belonging to the monocyte/macrophage lineage to both exogenous and endogenous peroxynitrite. The notion that two different pathways mediated by downstream products of arachidonic acid metabolism converge in Bad phosphorylation emphasizes the relevance of this strategy for the regulation of macrophage survival to peroxynitrite at the inflammatory sites.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/immunology , Dinoprostone/immunology , Macrophages, Peritoneal/immunology , Mitochondrial Proteins/immunology , Monocytes/immunology , Peroxynitrous Acid/immunology , Protein Kinase C-alpha/immunology , Signal Transduction/immunology , bcl-Associated Death Protein/immunology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/immunology , Animals , Cell Death/drug effects , Cell Death/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cyclic AMP/immunology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/metabolism , Dinoprostone/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Humans , Hydroxyeicosatetraenoic Acids/immunology , Hydroxyeicosatetraenoic Acids/metabolism , Inflammation/enzymology , Inflammation/immunology , Macrophages, Peritoneal/enzymology , Mitochondrial Proteins/metabolism , Monocytes/enzymology , Peroxynitrous Acid/metabolism , Phospholipase A2 Inhibitors , Phospholipases A2/immunology , Phospholipases A2/metabolism , Phosphorylation/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP2 Subtype , Signal Transduction/drug effects , U937 Cells , bcl-Associated Death Protein/metabolism
16.
J Exp Med ; 205(12): 2791-801, 2008 Nov 24.
Article in English | MEDLINE | ID: mdl-18955568

ABSTRACT

Virulent Mycobacterium tuberculosis (Mtb) induces a maladaptive cytolytic death modality, necrosis, which is advantageous for the pathogen. We report that necrosis of macrophages infected with the virulent Mtb strains H37Rv and Erdmann depends on predominant LXA(4) production that is part of the antiinflammatory and inflammation-resolving action induced by Mtb. Infection of macrophages with the avirulent H37Ra triggers production of high levels of the prostanoid PGE(2), which promotes protection against mitochondrial inner membrane perturbation and necrosis. In contrast to H37Ra infection, PGE(2) production is significantly reduced in H37Rv-infected macrophages. PGE(2) acts by engaging the PGE(2) receptor EP2, which induces cyclic AMP production and protein kinase A activation. To verify a role for PGE(2) in control of bacterial growth, we show that infection of prostaglandin E synthase (PGES)(-/-) macrophages in vitro with H37Rv resulted in significantly higher bacterial burden compared with wild-type macrophages. More importantly, PGES(-/-) mice harbor significantly higher Mtb lung burden 5 wk after low-dose aerosol infection with virulent Mtb. These in vitro and in vivo data indicate that PGE(2) plays a critical role in inhibition of Mtb replication.


Subject(s)
Cell Death/immunology , Dinoprostone/immunology , Immunity, Innate/physiology , Lipoxins/immunology , Macrophages/immunology , Mycobacterium tuberculosis , Receptors, Prostaglandin E/immunology , Animals , Cells, Cultured , Dinoprostone/chemistry , Dinoprostone/genetics , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Lipoxins/chemistry , Lipoxins/genetics , Lung/immunology , Lung/microbiology , Macrophages/cytology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/ultrastructure , Molecular Structure , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Prostaglandin-E Synthases , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Tuberculosis/immunology
17.
Allergol Int ; 57(3): 187-96, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18490861

ABSTRACT

The purpose of this review is to summarize the role of prostaglandins (PGs) in allergic inflammation and to know the value of PGs, as a target molecule for an anti-allergic drug. PGD(2) is the major PG produced by the cyclooxygenase pathway in mast cells. Our and others findings indicate that PGD(2) is one of the potent allergic inflammatory mediators and must be a target molecule of anti-allergic agent. From our data, one of PGD(2) receptor antagonists show clear inhibition of airway hypersensitivity caused by allergic reaction. Concerning the role of PGE(2) in allergic inflammation, conflicting results have been reported. Many experimental data suggest an individual role of each PGE(2) receptor, EP(1), EP(2), EP(3) and EP(4) in allergic reaction. Our results indicate the protective action of PGE(2) on allergic reaction via EP(3). In addition, one of EP(3) agonists clearly inhibits the allergic airway inflammation. These findings indicate the value of EP(3) agonists as an anti-allergic agent. In addition, some investigators including us reported that PGI(2) plays an important role for the protection of the onset of allergic reaction. However, the efficacy of PGI(2) analogue as an anti-allergic agent is not yet fully investigated. Finally, the role of thromboxane A(2) (TxA(2)) in allergic reaction is discussed. Our experimental results suggest a different participation of TxA(2) in allergic reaction of airway and skin. In this review, the role of PGs in allergic inflammation is summarized and the value of PGs as a target molecule for developing a new anti-allergic agent will be discussed.


Subject(s)
Anti-Allergic Agents/immunology , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Prostaglandins/immunology , Animals , Anti-Allergic Agents/pharmacology , Bronchial Provocation Tests , Disease Models, Animal , Humans , Mice , Receptors, Immunologic/agonists , Receptors, Immunologic/immunology , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/immunology , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E, EP3 Subtype , Thromboxane A2/immunology
18.
Am J Physiol Lung Cell Mol Physiol ; 295(1): L114-22, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18456799

ABSTRACT

Impaired host defense post-bone marrow transplant (BMT) is related to overproduction of prostaglandin E(2) (PGE(2)) by alveolar macrophages (AMs). We show AMs post-BMT overproduce granulocyte-macrophage colony-stimulating factor (GM-CSF), whereas GM-CSF in lung homogenates is impaired both at baseline and in response to infection post-BMT. Homeostatic regulation of GM-CSF may occur by hematopoietic/structural cell cross talk. To determine whether AM overproduction of GM-CSF influenced immunosuppression post-BMT, we compared mice that received BMT from wild-type donors (control BMT) or mice that received BMT from GM-CSF-/- donors (GM-CSF-/- BMT) with untransplanted mice. GM-CSF-/- BMT mice were less susceptible to pneumonia with Pseudomonas aeruginosa compared with control BMT mice and showed antibacterial responses equal to or better than untransplanted mice. GM-CSF-/- BMT AMs displayed normal phagocytosis and a trend toward enhanced bacterial killing. Surprisingly, AMs from GM-CSF-/- BMT mice overproduced PGE(2), but expression of the inhibitory EP(2) receptor was diminished. As a consequence of decreased EP(2) receptor expression, we found diminished accumulation of cAMP in response to PGE(2) stimulation in GM-CSF-/- BMT AMs compared with control BMT AMs. In addition, GM-CSF-/- BMT AMs retained cysteinyl leukotriene production and normal TNF-alpha response compared with AMs from control BMT mice. GM-CSF-/- BMT neutrophils also showed improved bacterial killing. Although genetic ablation of GM-CSF in hematopoietic cells post-BMT improved host defense, transplantation of wild-type bone marrow into GM-CSF-/- recipients demonstrated that parenchymal cell-derived GM-CSF is necessary for effective innate immune responses post-BMT. These results highlight the complex regulation of GM-CSF and innate immunity post-BMT.


Subject(s)
Bone Marrow Transplantation , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Macrophages, Alveolar/immunology , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Animals , Dinoprostone/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Immunity, Innate/genetics , Macrophages, Alveolar/pathology , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Phagocytosis/genetics , Phagocytosis/immunology , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/pathology , Pseudomonas Infections/genetics , Pseudomonas Infections/pathology , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E, EP2 Subtype
19.
Eur J Immunol ; 38(7): 1900-12, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18506884

ABSTRACT

Prostaglandin E(2) (PGE(2)) is a major mediator of inflammation and is present at high concentrations in the synovial fluid of rheumatoid arthritis (RA) patients. PGE(2), acting through the EP4 receptor, has both pro- and anti-inflammatory roles in vivo. To shed light on this dual role of PGE(2), we investigated its effects in whole blood and in primary human fibroblast-like synoviocytes (FLS). Gene expression analysis in human leukocytes, confirmed at the protein level, revealed an EP4-dependent inhibition of the expression of genes involved in the IFN-gamma-activation pathway, including IFN-gamma itself. This effect of the PGE(2)/EP4 axis on IFN-gamma is a reciprocal phenomenon since IFN-gamma blocks PGE(2) release and blocks EP receptor expression. The mutually antagonistic relationship between IFN-gamma and PGE(2) extends to downstream cytokine and chemokine release; PGE(2) counters the effects of IFN-gamma, on the release of IP-10, IL-8, TNF-alpha and IL-1beta. To gain further insight into IFN-gamma-mediated cellular events in RA, we assessed the effects of IFN-gamma on gene expression in FLS. We observed an IFN-gamma-dependent up-regulation of macrophage-attracting chemokines, and down-regulation of metalloprotease expression. These results suggest the existence of a mutually antagonistic relationship between PGE(2) and IFN-gamma, which may represent a fundamental mechanism of immune control in diseases such as RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Cytokines/metabolism , Dinoprostone/metabolism , Interferon-gamma/metabolism , Receptors, Prostaglandin E/metabolism , Arthritis, Rheumatoid/metabolism , Cytokines/immunology , Dinoprostone/blood , Down-Regulation , Gene Expression , Gene Regulatory Networks , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Oligonucleotide Array Sequence Analysis , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E, EP4 Subtype , Synovial Fluid/cytology , Synovial Fluid/immunology , Up-Regulation
20.
J Exp Med ; 204(12): 2865-74, 2007 Nov 26.
Article in English | MEDLINE | ID: mdl-17967902

ABSTRACT

Prostaglandin E2 (PGE2) exerts its actions via four subtypes of the PGE receptor, EP1-4. We show that mice deficient in EP1 exhibited significantly attenuated Th1 response in contact hypersensitivity induced by dinitrofluorobenzene (DNFB). This phenotype was recapitulated in wild-type mice by administration of an EP1-selective antagonist during the sensitization phase, and by adoptive transfer of T cells from sensitized EP1-/- mice. Conversely, an EP1-selective agonist facilitated Th1 differentiation of naive T cells in vitro. Finally, CD11c+ cells containing the inducible form of PGE synthase increased in number in the draining lymph nodes after DNFB application. These results suggest that PGE2 produced by dendritic cells in the lymph nodes acts on EP1 in naive T cells to promote Th1 differentiation.


Subject(s)
Receptors, Prostaglandin E/immunology , Th1 Cells/immunology , Animals , Antigen-Presenting Cells/immunology , Cell Differentiation , Cinnamates/pharmacology , Dendritic Cells/immunology , Dendritic Cells/physiology , Dinoprostone/physiology , Lymph Nodes/immunology , Lymph Nodes/physiology , Mice , Mice, Knockout , Prostaglandins/physiology , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/deficiency , Receptors, Prostaglandin E, EP1 Subtype , T-Lymphocyte Subsets/immunology , Th1 Cells/cytology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...