Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Front Immunol ; 11: 1224, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32636842

RESUMO

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.


Assuntos
Asma/metabolismo , Dinoprostona/metabolismo , Switching de Imunoglobulina/imunologia , Imunoglobulina E/biossíntese , PPAR gama/metabolismo , Animais , Asma/imunologia , Dinoprostona/imunologia , Imunoglobulina E/imunologia , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/imunologia , Estabilidade Proteica , Receptores de Prostaglandina E Subtipo EP4/deficiência , Receptores de Prostaglandina E Subtipo EP4/imunologia , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais/imunologia
2.
J Allergy Clin Immunol ; 146(6): 1387-1396.e13, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32407837

RESUMO

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.


Assuntos
Anafilaxia/imunologia , Dinoprostona/deficiência , Mastócitos/imunologia , Anafilaxia/patologia , Animais , Dinoprostona/imunologia , Suscetibilidade a Doenças/imunologia , MAP Quinases Reguladas por Sinal Extracelular/imunologia , Humanos , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fosfolipase C gama/imunologia , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , Índice de Gravidade de Doença
3.
Sci Rep ; 8(1): 13351, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30190508

RESUMO

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.


Assuntos
Dinoprostona/imunologia , Tolerância Imunológica , Linfócitos/imunologia , Miocárdio/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , Células-Tronco/imunologia , Animais , Comunicação Celular/imunologia , Proliferação de Células , Cães , Antígenos de Histocompatibilidade Classe I/imunologia , Linfócitos/citologia , Miocárdio/citologia , Células-Tronco/citologia
4.
Proc Natl Acad Sci U S A ; 115(36): E8469-E8478, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30127026

RESUMO

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.


Assuntos
Citrobacter rodentium/imunologia , Colite/imunologia , Células Dendríticas/imunologia , Dinoprostona/imunologia , Infecções por Enterobacteriaceae/imunologia , Intestinos/imunologia , Macrófagos/imunologia , Animais , Colite/microbiologia , Colite/patologia , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Feminino , Intestinos/microbiologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Receptores de Prostaglandina E Subtipo EP4/imunologia
5.
JCI Insight ; 3(3)2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29415894

RESUMO

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.


Assuntos
Autoimunidade/efeitos dos fármacos , Diabetes Mellitus Tipo 1/imunologia , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Compostos de Sulfonilureia/administração & dosagem , Células Th1/imunologia , Administração Oral , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Dinoprostona/imunologia , Dinoprostona/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Interleucina-1/imunologia , Interleucina-1/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/imunologia , Receptor de Interferon alfa e beta/metabolismo , Receptores de Prostaglandina E Subtipo EP4/imunologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células Th1/efeitos dos fármacos , Células Th1/metabolismo
6.
Front Immunol ; 9: 2954, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619314

RESUMO

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.


Assuntos
Colite/imunologia , Dinoprostona/imunologia , Prostaglandina-E Sintases/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , Linfócitos T/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Colite/genética , Colite/metabolismo , Dinoprostona/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Prostaglandina-E Sintases/genética , Prostaglandina-E Sintases/metabolismo , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
8.
Diabetes ; 66(2): 494-504, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27815317

RESUMO

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.


Assuntos
Ciclo-Oxigenase 2/imunologia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/imunologia , Nefropatias Diabéticas/imunologia , Macrófagos/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , Albuminúria , Animais , Células Cultivadas , Ciclo-Oxigenase 2/genética , Fibrose , Immunoblotting , Imuno-Histoquímica , Rim/metabolismo , Rim/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , NF-kappa B/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos , Óxido Nítrico Sintase Tipo II/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Prostaglandina E/imunologia , Transdução de Sinais , Linfócitos T/imunologia
9.
Science ; 351(6279): 1333-8, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26989254

RESUMO

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.


Assuntos
Dinoprostona/imunologia , Inflamação/imunologia , Interleucinas/imunologia , Intestinos/imunologia , Linfócitos/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Expressão Gênica , Humanos , Imunidade Inata , Inflamação/tratamento farmacológico , Inflamação/microbiologia , Intestinos/microbiologia , Camundongos , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/genética , Transdução de Sinais , Interleucina 22
10.
Nat Commun ; 7: 10760, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26907476

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Dinoprostona/farmacologia , Interleucina-1beta/efeitos dos fármacos , Leucotrieno B4/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Picadas de Escorpião/imunologia , Venenos de Escorpião/farmacologia , Animais , Araquidonato 5-Lipoxigenase/genética , Western Blotting , Proteínas de Transporte/imunologia , Celecoxib/farmacologia , AMP Cíclico/imunologia , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/imunologia , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/imunologia , Técnicas In Vitro , Indóis/farmacologia , Indometacina/farmacologia , Inflamassomos/imunologia , Interleucina-1beta/imunologia , Leucotrieno B4/imunologia , Inibidores de Lipoxigenase/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Knockout , NF-kappa B/efeitos dos fármacos , NF-kappa B/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fosfoproteínas , Antagonistas de Prostaglandina/farmacologia , Receptores de Prostaglandina E Subtipo EP2/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP4/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP4/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Picadas de Escorpião/mortalidade , Escorpiões , Xantonas/farmacologia
11.
PLoS One ; 11(1): e0147721, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26824354

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/imunologia , Anti-Inflamatórios não Esteroides/farmacologia , Curcumina/farmacologia , Dinoprostona/imunologia , Microglia/efeitos dos fármacos , Microglia/imunologia , Fragmentos de Peptídeos/imunologia , Fagocitose/efeitos dos fármacos , Animais , AMP Cíclico/imunologia , Proteínas Quinases Dependentes de AMP Cíclico/imunologia , Camundongos , Microglia/citologia , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , Transdução de Sinais/efeitos dos fármacos
12.
Cell Mol Immunol ; 13(2): 240-50, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26189370

RESUMO

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.


Assuntos
Antígenos CD40/imunologia , Células Dendríticas/imunologia , Dinoprostona/imunologia , Subunidade p19 da Interleucina-23/imunologia , Subunidade p50 de NF-kappa B/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , Transdução de Sinais/imunologia , Animais , Antígenos CD40/genética , Dinoprostona/genética , Feminino , Subunidade p19 da Interleucina-23/genética , Camundongos , Camundongos Mutantes , Subunidade p50 de NF-kappa B/genética , Receptores de Prostaglandina E Subtipo EP4/genética , Transdução de Sinais/genética , Transcrição Gênica/imunologia
13.
Int Immunopharmacol ; 28(1): 376-81, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26122137

RESUMO

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.


Assuntos
Criptococose/imunologia , Cryptococcus neoformans , Dinoprostona/antagonistas & inibidores , Macrófagos Alveolares/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Criptococose/microbiologia , Citocinas/genética , Citocinas/imunologia , Dinoprostona/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Camundongos Endogâmicos BALB C , Camundongos Knockout , Antagonistas de Prostaglandina/farmacologia , RNA Mensageiro/metabolismo , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/imunologia , Xantonas/farmacologia
14.
J Allergy Clin Immunol ; 134(5): 1163-74.e16, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24948368

RESUMO

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.


Assuntos
Asma Induzida por Aspirina/imunologia , Tolerância Imunológica , Células Mieloides/imunologia , Transdução de Sinais/imunologia , Alérgenos/toxicidade , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/farmacologia , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Arginase/genética , Arginase/imunologia , Aspirina/efeitos adversos , Aspirina/farmacologia , Asma Induzida por Aspirina/genética , Asma Induzida por Aspirina/patologia , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/imunologia , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/imunologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Células Mieloides/patologia , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células Th2/imunologia , Células Th2/patologia
15.
Nat Commun ; 4: 2657, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24150174

RESUMO

Although it has long been known that patients with sepsis often have thrombocytopenia and that septic patients with severe thrombocytopenia have a poor prognosis and higher mortality, the role of platelets in the pathogenesis of sepsis is poorly understood. Here we report a protective role of platelets in septic shock. We show that experimental thrombocytopenia induced by intraperitoneal injection of an anti-glycoprotein Ibα monoclonal antibody increases mortality and aggravates organ failure, whereas transfusion of platelets reduces mortality in lipopolysaccharide-induced endotoxemia and a bacterial infusion mouse sepsis model. Plasma concentrations of proinflammatory cytokines TNF-α and IL-6 are elevated by thrombocytopenia and decreased by platelet transfusion in septic mice. Furthermore, we identify that platelets protect from septic shock by inhibiting macrophage-dependent inflammation via the COX1/PGE2/EP4-dependent pathway. Thus, these findings demonstrate a previously unappreciated role for platelets in septic shock and suggest that platelet transfusion may be effective in treating severely septic patients.


Assuntos
Plaquetas/imunologia , Macrófagos Peritoneais/imunologia , Transfusão de Plaquetas , Choque Séptico/terapia , Trombocitopenia/prevenção & controle , Animais , Anticorpos Anti-Idiotípicos/farmacologia , Plaquetas/citologia , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/imunologia , Dinoprostona/genética , Dinoprostona/imunologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Inflamação/prevenção & controle , Interleucina-6/biossíntese , Interleucina-6/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Complexo Glicoproteico GPIb-IX de Plaquetas/imunologia , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/imunologia , Choque Séptico/induzido quimicamente , Choque Séptico/genética , Choque Séptico/patologia , Transdução de Sinais , Trombocitopenia/induzido quimicamente , Trombocitopenia/genética , Trombocitopenia/patologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
16.
Mol Immunol ; 54(3-4): 284-95, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23337716

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Dinoprostona/imunologia , Monócitos/imunologia , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , AMP Cíclico/imunologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/imunologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Dinoprostona/metabolismo , Fatores de Troca do Nucleotídeo Guanina/imunologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Subunidade p40 da Interleucina-12/imunologia , Subunidade p40 da Interleucina-12/metabolismo , Subunidade p19 da Interleucina-23/imunologia , Subunidade p19 da Interleucina-23/metabolismo , Lipopolissacarídeos/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Prostaglandina E Subtipo EP2/biossíntese , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/biossíntese , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo
17.
Prostaglandins Other Lipid Mediat ; 100-101: 30-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23237743

RESUMO

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.


Assuntos
Adenosina/farmacologia , Dinoprostona/farmacologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Adenosina/uso terapêutico , Análise de Variância , Animais , Dinoprostona/uso terapêutico , Sinergismo Farmacológico , Quimioterapia Combinada , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/imunologia , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptor A2A de Adenosina/imunologia , Receptor A2A de Adenosina/metabolismo , Receptores de Prostaglandina E Subtipo EP4/imunologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo
18.
Eur J Pharmacol ; 699(1-3): 141-9, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23220160

RESUMO

The cyclooxygenase (COX) pathway and prostanoids may critically contribute to the early allergic airway response. In the rat lung, serotonin (5-HT) is a major mediator of antigen-induced contractions. The aim of this study was therefore to examine the relative role of the COX pathway and serotonin for antigen-induced contractions in the rat lung. Airway responses were studied in rat precision-cut lung slices (PCLS). Lung slices were stimulated with ovalbumin or serotonin after pretreatment with COX inhibitors or specific TP or EP receptor antagonists. Changes in airway size (contractions/relaxations) were measured by a digital video camera. The supernatants were analysed for changes in prostaglandin and serotonin release. Airway contractions to ovalbumin were attenuated by the unselective COX inhibitor indomethacin, the selective COX-1 inhibitor FR-122047 and COX-2 inhibitor celecoxib. The EP(1) receptor antagonist ONO-8713 reduced the contractions, whereas the EP(4) receptor antagonist L-161,982 significantly increased the contractile response to ovalbumin. The 5-HT(2A) receptor antagonist ketanserin completely inhibited the ovalbumin-induced contractions. The different COX inhibitors decreased the production of prostaglandins but did not affect the synthesis of serotonin. The serotonin-induced bronchoconstriction was attenuated by celecoxib and ONO-8713, but not by methacholine. Taken together, our data indicate that PGE(2) is the main prostanoid involved in the early allergic airway response in the rat lung. PGE(2) appears to act both as a primary mediator of antigen-induced airway contraction via the EP(4) receptor and as a downstream modulator of serotonin-induced bronchoconstriction via the EP(1) receptor.


Assuntos
Dinoprostona/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Serotonina/metabolismo , Animais , Antígenos/imunologia , Broncoconstrição/imunologia , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/imunologia , Pulmão/imunologia , Ovalbumina/imunologia , Prostaglandina-Endoperóxido Sintases/imunologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Ratos , Ratos Wistar , Receptores de Prostaglandina E Subtipo EP1/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia
19.
Shock ; 38(6): 620-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23143054

RESUMO

The mechanism of immunosuppression induced by severe sepsis is not fully understood. The production of prostaglandin E2 (PGE2) during sepsis is well known, but its role in long-term consequences of sepsis has not been explored. The current study evaluates the role of PGE2 in the development of immunosuppression secondary to sepsis and its potential as therapeutic target. Cecal ligation and puncture was used as an experimental model for sepsis induction in Balb/c and C57BL/6 mice. Immunosuppression was evaluated by the response to secondary infection with Aspergillus fumigatus in sepsis survivors. The role of prostanoids was evaluated in vivo and in vitro by treatment with the cyclooxygenase inhibitor ketoprofen. Balb/c mice were more susceptible than C57BL/6 to severe sepsis and to secondary infection, with a greater mortality rate. Prostaglandin E2 concentrations found in bronchoalveolar lavage in sham and cecal ligation and puncture group after fungal challenge were much higher in Balb/c than in C57BL/6 mice. Ketoprofen treatment improved survival of septic Balb/c mice subjected to secondary infection, while also enhancing macrophage phagocytosis and neutrophil recruitment to the lungs. We identified a pivotal role for PGE2 acting on EP4 receptors in modulating cytokine production differentially by sham and septic macrophages. Furthermore, sepsis also altered key enzymes in PGE2 synthesis and degradation. Our results indicate the involvement of PGE2 in severe sepsis-induced immunosuppression. Inhibition of PGE2 production represents an attractive target to improve innate immune response against secondary infection in the immunocompromised host.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Dinoprostona/imunologia , Tolerância Imunológica/efeitos dos fármacos , Cetoprofeno/efeitos adversos , Sepse/imunologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Citocinas/imunologia , Modelos Animais de Doenças , Cetoprofeno/farmacologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Receptores de Prostaglandina E Subtipo EP4/imunologia , Sepse/tratamento farmacológico , Índice de Gravidade de Doença
20.
J Immunol ; 189(10): 4777-86, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23066152

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Células Dendríticas/imunologia , Tolerância Imunológica/fisiologia , Monócitos/imunologia , Sêmen/imunologia , Adulto , Antígenos CD1/imunologia , Diferenciação Celular/efeitos dos fármacos , Citocinas/imunologia , Células Dendríticas/citologia , Feminino , Humanos , Tolerância Imunológica/efeitos dos fármacos , Receptores de Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Masculino , Monócitos/citologia , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA