Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunology ; 172(1): 144-162, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38361249

RESUMO

Macrophages expressing group V phospholipase A2 (Pla2g5) release the free fatty acid (FFA) linoleic acid (LA), potentiating lung type 2 inflammation. Although Pla2g5 and LA increase in viral infections, their role remains obscure. We generated Pla2g5flox/flox mice, deleted Pla2g5 by using the Cx3cr1cre transgene, and activated bone marrow-derived macrophages (BM-Macs) with poly:IC, a synthetic double-stranded RNA that triggers a viral-like immune response, known Pla2g5-dependent stimuli (IL-4, LPS + IFNγ, IL-33 + IL-4 + GM-CSF) and poly:IC + LA followed by lipidomic and transcriptomic analysis. Poly:IC-activated Pla2g5flox/flox;Cx3cr1cre/+ BM-Macs had downregulation of major bioactive lipids and critical enzymes producing those bioactive lipids. In addition, AKT phosphorylation was lower in poly:IC-stimulated Pla2g5flox/flox;Cx3cr1cre/+ BM-Macs, which was not restored by adding LA to poly:IC-stimulated BM-Macs. Consistently, Pla2g5flox/flox;Cx3cr1cre/+ mice had diminished poly:IC-induced lung inflammation, including inflammatory macrophage proliferation, while challenging Pla2g5flox/flox;Cx3cr1cre/+ mice with poly:IC + LA partially restored lung inflammation and inflammatory macrophage proliferation. Finally, mice lacking FFA receptor-1 (Ffar1)-null mice had reduced poly:IC-induced lung cell recruitment and tissue macrophage proliferation, not corrected by LA. Thus, Pla2g5 contributes to poly:IC-induced lung inflammation by regulating inflammatory macrophage proliferation and LA/Ffar1-mediated lung cell recruitment and tissue macrophage proliferation.


Assuntos
Ácido Linoleico , Pneumonia , Animais , Camundongos , Proliferação de Células , Interleucina-4/metabolismo , Ácido Linoleico/metabolismo , Pulmão , Macrófagos
2.
Front Cell Dev Biol ; 10: 966950, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36105351

RESUMO

Increasing evidence points to the involvement of group IIA secreted phospholipase A2 (sPLA2-IIA) in pathologies characterized by abnormal osteoclast bone-resorption activity. Here, the role of this moonlighting protein has been deepened in the osteoclastogenesis process driven by the RANKL cytokine in RAW264.7 macrophages and bone-marrow derived precursor cells from BALB/cJ mice. Inhibitors with distinct selectivity toward sPLA2-IIA activities and recombinant sPLA2-IIA (wild-type or catalytically inactive forms, full-length or partial protein sequences) were instrumental to dissect out sPLA2-IIA function, in conjunction with reduction of sPLA2-IIA expression using small-interfering-RNAs and precursor cells from Pla2g2a knock-out mice. The reported data indicate sPLA2-IIA participation in murine osteoclast maturation, control of syncytium formation and resorbing activity, by mechanisms that may be both catalytically dependent and independent. Of note, these studies provide a more complete understanding of the still enigmatic osteoclast multinucleation process, a crucial step for bone-resorbing activity, uncovering the role of sPLA2-IIA interaction with a still unidentified receptor to regulate osteoclast fusion through p38 SAPK activation. This could pave the way for the design of specific inhibitors of sPLA2-IIA binding to interacting partners implicated in osteoclast syncytium formation.

4.
Trends Parasitol ; 38(3): 191-192, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35078723

RESUMO

Type 2 inflammation (T2I) accompanies many inflammatory diseases. In a recent issue of Cell, Ahrends et al. demonstrate that helminth-elicited T2I preserves excitatory neurons and enteric function through the expansion of Arginase-1 (Arg-1)-expressing macrophages, thereby extending our understanding of the protective functions that T2I can orchestrate in inflamed barrier tissue.


Assuntos
Inflamação , Macrófagos , Humanos
5.
Biomedicines ; 9(5)2021 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-34064389

RESUMO

BACKGROUND: Pulmonary macrophages are a highly heterogeneous cell population distributed in different lung compartments. METHODS: We separated two subpopulations of macrophages from human lung parenchyma according to flotation over density gradients. RESULTS: Two-thirds 65.4% of the lung macrophages have a density between 1.065 and 1.078 (high-density macrophages: HDMs), and the remaining one-third (34.6) had a density between 1.039 and 1.052 (low-density macrophages: LDMs). LDMs had a larger area (691 vs. 462 µm2) and cell perimeter (94 vs. 77 µm) compared to HDMs. A significantly higher percentage of HDMs expressed CD40, CD45, and CD86 compared to LDMs. In contrast, a higher percentage of LDMs expressed the activation markers CD63 and CD64. The release of TNF-α, IL-6, IL-10 and IL-12 induced by lipopolysaccharide (LPS) was significantly higher in HDMs than in LDMs. CONCLUSION: The human lung contains two subpopulations of macrophages that differ in buoyancy, morphometric parameters, surface marker expression and response to LPS. These subpopulations of macrophages probably play distinct roles in lung inflammation and immune responses.

6.
Molecules ; 27(1)2021 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-35011385

RESUMO

Macrophages have diverse functions in the pathogenesis, resolution, and repair of inflammatory processes. Elegant studies have elucidated the metabolomic and transcriptomic profiles of activated macrophages. However, the versatility of macrophage responses in inflammation is likely due, at least in part, to their ability to rearrange their repertoire of bioactive lipids, including fatty acids and oxylipins. This review will describe the fatty acids and oxylipins generated by macrophages and their role in type 1 and type 2 immune responses. We will highlight lipidomic studies that have shaped the current understanding of the role of lipids in macrophage polarization.


Assuntos
Ácidos Graxos/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Oxilipinas/metabolismo , Animais , Biomarcadores , Citocinas/metabolismo , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Humanos , Imunidade , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação , Metabolismo dos Lipídeos , Lipidômica/métodos , Redes e Vias Metabólicas , Transdução de Sinais
8.
Biomolecules ; 11(1)2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33383652

RESUMO

Macrophages activated by Interleukin (IL)-4 (M2) or LPS+ Interferon (IFN)γ (M1) perform specific functions respectively in type 2 inflammation and killing of pathogens. Group V phospholipase A2 (Pla2g5) is required for the development and functions of IL-4-activated macrophages and phagocytosis of pathogens. Pla2g5-generated bioactive lipids, including lysophospholipids (LysoPLs), fatty acids (FAs), and eicosanoids, have a role in many diseases. However, little is known about their production by differentially activated macrophages. We performed an unbiased mass-spectrometry analysis of phospholipids (PLs), LysoPLs, FAs, and eicosanoids produced by Wild Type (WT) and Pla2g5-null IL-4-activated bone marrow-derived macrophages (IL-4)BM-Macs (M2) and (LPS+IFNγ)BM-Macs (M1). Phosphatidylcholine (PC) was preferentially metabolized in (LPS+IFNγ)BM-Macs and Phosphatidylethanolamine (PE) in (IL-4)BM-Macs, with Pla2g5 contributing mostly to metabolization of selected PE molecules. While Pla2g5 produced palmitic acid (PA) in (LPS+IFNγ)BM-Macs, the absence of Pla2g5 increased myristic acid (MA) in (IL-4)BM-Macs. Among eicosanoids, Prostaglandin E2 (PGE2) and prostaglandin D2 (PGD2) were significantly reduced in (IL-4)BM-Macs and (LPS+IFNγ)BM-Macs lacking Pla2g5. Instead, the IL-4-induced increase in 20-carboxy arachidonic acid (20CooH AA) was dependent on Pla2g5, as was the production of 12-hydroxy-heptadecatrienoic acid (12-HHTrE) in (LPS+IFNγ)BM-Macs. Thus, Pla2g5 contributes to PE metabolization, PGE2 and PGD2 production independently of the type of activation, while in (IL-4)BM-Macs, Pla2g5 regulates selective lipid pathways and likely novel functions.


Assuntos
Fosfolipases A2 do Grupo V/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Fosfolipídeos/imunologia , Animais , Células Cultivadas , Humanos , Inflamação/imunologia , Interleucina-4/imunologia , Camundongos , Fosfolipídeos/análise
9.
J Clin Invest ; 129(12): 5169-5186, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31638598

RESUMO

Antagonists of the type 1 cysteinyl leukotriene receptor (CysLT1R) are widely used to treat asthma and allergic rhinitis, with variable response rates. Alveolar macrophages express UDP-specific P2Y6 receptors that can be blocked by off-target effects of CysLT1R antagonists. Sensitizing intranasal doses of an extract from the house dust mite Dermatophagoides farinae (Df) sharply increased the levels of UDP detected in bronchoalveolar lavage fluid of mice. Conditional deletion of P2Y6 receptors before sensitization exacerbated eosinophilic lung inflammation and type 2 cytokine production in response to subsequent Df challenge. P2Y6 receptor signaling was necessary for dectin-2-dependent production of protective IL-12p40 and Th1 chemokines by alveolar macrophages, leading to activation of NK cells to generate IFN-γ. Administration of CysLT1R antagonists during sensitization blocked UDP-elicited potentiation of IL-12p40 production by macrophages in vitro, suppressed the Df-induced production of IL-12p40 and IFN-γ in vivo, and suppressed type 2 inflammation only in P2Y6-deficient mice. Thus, P2Y6 receptor signaling drives an innate macrophage/IL-12/NK cell/IFN-γ axis that prevents inappropriate allergic type 2 immune responses on respiratory allergen exposure and counteracts the Th2 priming effect of CysLT1R signaling at sensitization. Targeting P2Y6 signaling might prove to be a potential additional treatment strategy for allergy.


Assuntos
Hipersensibilidade/metabolismo , Inflamação/metabolismo , Leucotrienos/metabolismo , Macrófagos Alveolares/metabolismo , Receptores Purinérgicos P2/metabolismo , Transdução de Sinais , Alérgenos/imunologia , Animais , Antígenos de Dermatophagoides/imunologia , Asma/imunologia , Bioensaio , Líquido da Lavagem Broncoalveolar , Linfócitos T CD8-Positivos/citologia , Dermatophagoides farinae , Feminino , Células-Tronco Hematopoéticas/citologia , Subunidade p35 da Interleucina-12/metabolismo , Ligantes , Pulmão/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Eosinofilia Pulmonar
10.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(6): 819-826, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30308324

RESUMO

Group V Phospholipase A2 (Pla2g5) is a member of the PLA2 family of lipid-generating enzymes. It is expressed in immune and non-immune cell types and is inducible during several pathologic conditions serving context-specific functions. In this review, we recapitulate the protective and detrimental functions of Pla2g5 investigated through preclinical and translational approaches. This article is part of a Special Issue entitled Novel functions of phospholipase A2 Guest Editors: Makoto Murakami and Gerard Lambeau.


Assuntos
Fosfolipases A2/metabolismo , Animais , Humanos , Imunidade/imunologia , Fosfolipases A2/imunologia
11.
J Biol Chem ; 292(20): 8195-8206, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28341741

RESUMO

When activated through toll-like receptors (TLRs), macrophages generate IL-33, an IL-1 family member that induces innate immune responses through ST2 signaling. LPS, a TLR4 ligand, induces macrophages to generate prostaglandin E2 (PGE2) through inducible COX-2 and microsomal PGE2 synthase 1 (mPGES-1) (1). We demonstrate that IL-33 production by bone marrow-derived murine macrophages (bmMFs) requires the generation of endogenous PGE2 and the intrinsic expression of EP2 receptors to amplify NF-κB-dependent, LPS-induced IL-33 expression via exchange protein activated by cAMP (EPAC). Compared with WT cells, bmMFs lacking either mPGES-1 or EP2 receptors displayed reduced LPS-induced IL-33 levels. A selective EP2 agonist and, to a lesser extent, EP4 receptor agonist potentiated LPS-induced IL-33 generation from both mPGES-1-null and WT bmMFs, whereas EP1 and EP3 receptor agonists were inactive. The effects of PGE2 depended on cAMP, were mimicked by an EPAC-selective agonist, and were attenuated by EPAC-selective antagonism and knockdown. LPS-induced p38 MAPK and NF-κB activations were necessary for both IL-33 production and PGE2 generation, and exogenous PGE2 partly reversed the suppression of IL-33 production caused by p38 MAPK and NF-κB inhibition. Mice lacking mPGES-1 showed lower IL-33 levels and attenuated lung inflammation in response to repetitive Alternaria inhalation challenges. Cumulatively, our data demonstrate that endogenous PGE2, EP2 receptors, and EPAC are prerequisites for maximal LPS-induced IL-33 expression and that exogenous PGE2 can amplify IL-33 production via EP2 and EP4 receptors. The ubiquitous induction of mPGES-1-dependent PGE2 may be crucial for innate immune system activation during various IL-33 driven pathologic disorders.


Assuntos
AMP Cíclico/metabolismo , Dinoprostona , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Interleucina-33/metabolismo , Macrófagos/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Animais , AMP Cíclico/genética , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Fatores de Troca do Nucleotídeo Guanina/genética , Interleucina-33/genética , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Mutantes , Prostaglandina-E Sintases/genética , Prostaglandina-E Sintases/metabolismo , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP4/genética
12.
PLoS One ; 11(7): e0158893, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27391897

RESUMO

Anion exchanger 2 (Ae2; gene symbol, Slc4a2) is a plasma membrane Cl-/HCO3- exchanger expressed in the gastrointestinal tract, kidney and bone. We have previously shown that Ae2 is required for the function of osteoclasts, bone resorbing cells of the macrophage lineage, to maintain homeostatic cytoplasmic pH and electroneutrality during acid secretion. Macrophages require endosomal acidification for pathogen killing during the process known as phagocytosis. Chloride is thought to be the principal ion responsible for maintaining electroneutrality during organelle acidification, but whether Cl-/HCO3- exchangers such as Ae2 contribute to macrophage function is not known. In this study we investigated the role of Ae2 in primary macrophages during phagocytosis. We find that Ae2 is expressed in macrophages where it regulates intracellular pH and the binding of Zymosan, a fungal cell wall derivative. Surprisingly, the transcription and surface expression of Dectin-1, the major phagocytic receptor for Candida albicans (C. albicans) and Zymosan, is reduced in the absence of Ae2. As a consequence, Zymosan-induced Tnfα expression is also impaired in Ae2-deficient macrophages. Similar to Ae2 deficiency, pharmacological alkalinization of lysosomal pH with bafilomycin A decreases both Dectin-1 mRNA and cell surface expression. Finally, Ae2-deficient macrophages demonstrate defective phagocytosis and killing of the human pathogenic fungus C. albicans. Our results strongly suggest that Ae2 is a critical factor in the innate response to C. albicans. This study represents an important contribution to a better understanding of how Dectin-1 expression and fungal clearance is regulated.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Lectinas Tipo C/imunologia , Macrófagos Peritoneais/imunologia , Fagocitose/imunologia , Animais , Candidíase/genética , Antiportadores de Cloreto-Bicarbonato/genética , Antiportadores de Cloreto-Bicarbonato/imunologia , Lectinas Tipo C/genética , Camundongos , Camundongos Knockout
13.
J Leukoc Biol ; 100(1): 131-41, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26936936

RESUMO

Phospholipases A2 are enzymes that liberate membrane-bound lipids in a tissue and cell-specific fashion. Group V secretory phospholipase A2 is necessary for the development of M2 macrophages and their effector functions in a mouse model of the T-helper-2 allergic airway inflammation. However, the function of group V phospholipase A2 in human M2 activation and T-helper-2 inflammation is ill-defined. Transglutaminase-2, a protein cross-linking enzyme, is a newly identified marker of both human and mouse interleukin-4-activated M2 macrophages and is also found in the lungs of patients with asthma. We report that group V phospholipase A2 and transglutaminase-2 colocalized in macrophages of human nasal polyp tissue obtained from patients with T-helper-2 eosinophilic inflammation, and their coexpression positively correlated with the number of eosinophils in each tissue specimen. We demonstrate that in human monocyte-derived macrophages activated by interleukin-4, group V phospholipase A2 translocated and colocalized with transglutaminase-2 in the cytoplasm and on the membrane of macrophages. Moreover, knocking down group V phospholipase A2 with small interfering ribonucleic acid reduced macrophage transglutaminase activity, whereas mass spectrometry analysis of lipids also showed reduced prostaglandin E2 production. Finally, exogenous prostaglandin E2 restored transglutaminase activity of group V phospholipase A2-small interfering ribonucleic acid-treated macrophages. Thus, our study shows a novel function of group V phospholipase A2 in regulating the transglutaminase activity of human interleukin-4-activated M2 macrophages through prostaglandin E2 generation and suggests that group V phospholipase A2 is a functionally relevant enzyme that may have therapeutic value for the treatment of human T-helper-2 inflammatory disorders.


Assuntos
Dinoprostona/metabolismo , Fosfolipases A2 do Grupo V/metabolismo , Inflamação/patologia , Interleucina-4/farmacologia , Macrófagos/patologia , Pólipos Nasais/metabolismo , Transglutaminases/metabolismo , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Células Cultivadas , Eosinófilos/imunologia , Eosinófilos/metabolismo , Eosinófilos/patologia , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Pessoa de Meia-Idade , Pólipos Nasais/patologia , Adulto Jovem
14.
J Immunol ; 190(12): 5927-38, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23650617

RESUMO

We reported that Pla2g5-null mice lacking group V secretory phospholipase A2 (gV-sPLA2) showed reduced eosinophilic pulmonary inflammation and Th2 cytokine generation when challenged with an extract from house dust mite Dermatophagoides farinae, compared with wild-type (WT) controls. Adoptive transfer studies suggested that gV-sPLA2 in dendritic cells was necessary for sensitization of Pla2g5-null mice, but was not sufficient to induce the effector phase of pulmonary inflammation. In this study, we demonstrate that gV-sPLA2 is inducibly expressed in mouse and human macrophages (M) activated by IL-4 and is required for the acquisition of M effector functions that facilitate the effector phase of pulmonary inflammation. We demonstrate that gV-sPLA2 expression in M is sufficient for the development of pulmonary inflammation, even when inflammation is induced by intrapulmonary administration of IL-4. The concentrations of CCL22/CCL17 and effector T cell recruitment are severely impaired in Pla2g5-null mice. Intratracheal transfers of enriched CD68(+) cells isolated from the lungs of D. farinae-challenged WT donor mice induce eosinophilia, chemokine production, and recruitment of T cells into the lungs of Pla2g5-null recipients previously sensitized by WT D. farinae-loaded dendritic cells. Our studies identified a unique function of gV-sPLA2 in activation of M and in their capacity to recruit T cells to amplify the effector phase of pulmonary inflammation.


Assuntos
Fosfolipases A2 do Grupo V/imunologia , Hipersensibilidade/imunologia , Ativação de Macrófagos/imunologia , Pneumonia/imunologia , Animais , Imunofluorescência , Fosfolipases A2 do Grupo V/metabolismo , Humanos , Hipersensibilidade/metabolismo , Imuno-Histoquímica , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
15.
J Immunol ; 187(3): 1486-95, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21724990

RESUMO

We show that the P2Y(6) receptor, a purinergic G protein-coupled receptor with a high affinity for the nucleotide uridine diphosphate, is an important endogenous inhibitor of T cell function in allergic pulmonary inflammation. Mice conditionally deficient in P2Y(6) receptors [p2ry6 (flox/flox);cre/+ mice] exhibited severe airway and tissue pathology relative to P2Y(6)-sufficient [p2ry6 (flox/flox)] littermates (+/+ mice) when treated intranasally with an extract of the dust mite Dermatophagoides farinae (Df). P2Y(6) receptors were inducibly expressed by lung, lymph node, and splenic CD4(+) and CD8(+) T cells of Df-treated +/+ mice. Df-restimulated P2Y(6)-deficient lymph node cells produced higher levels of Th1 and Th2 cytokines, and polyclonally stimulated P2Y(6)-deficient CD4(+) T cells proliferated faster than comparably stimulated P2Y(6)-sufficient cells. The absence of P2Y(6) receptors on CD4(+) cells, but not APCs, was sufficient to amplify cytokine generation. Thus, P2Y(6) receptors protect the lung against exuberant allergen-induced pulmonary inflammation by inhibiting the activation of effector T cells.


Assuntos
Alérgenos/imunologia , Antígenos de Dermatophagoides/imunologia , Linfócitos T CD4-Positivos/imunologia , Tolerância Imunológica , Ativação Linfocitária/imunologia , Receptores Purinérgicos P2/fisiologia , Hipersensibilidade Respiratória/imunologia , Alérgenos/administração & dosagem , Animais , Antígenos de Dermatophagoides/administração & dosagem , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Feminino , Tolerância Imunológica/genética , Ativação Linfocitária/genética , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores Purinérgicos P2/deficiência , Hipersensibilidade Respiratória/metabolismo , Hipersensibilidade Respiratória/patologia
16.
J Immunol ; 185(7): 4430-8, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20817863

RESUMO

We have previously shown that group V secretory phospholipase A(2) (sPLA(2)) regulates phagocytosis of zymosan and Candida albicans by a mechanism that depends on fusion of phagosomes with late endosomes in macrophages. In this study, we report that group V sPLA(2) (Pla2g5)-null mice exposed to an extract of house dust mite Dermatophagoides farinae had markedly reduced pulmonary inflammation and goblet cell metaplasia compared with wild-type (WT) mice. Pla2g5-null mice had also impaired Th2-type adaptive immune responses to D. farinae compared with WT mice. Pla2g5-null bone marrow-derived dendritic cells (BMDCs) activated by D. farinae had delayed intracellular processing of allergen and impaired allergen-dependent maturation, a pattern recapitulated by the native lung DCs of D. farinae-challenged mice. Adoptively transferred D. farinae-loaded Pla2g5-null BMDCs were less able than D. farinae-loaded WT BMDCs to induce pulmonary inflammation and Th2 polarization in WT mice. However, Pla2g5-null recipients transferred with WT or Pla2g5-null D. farinae-loaded BMDCs exhibited significantly reduced local inflammatory responses to D. farinae, even though the transfer of WT BMDCs still induced an intact Th2 cytokine response in regional lymph nodes. Thus, the expression of group V sPLA(2) in APCs regulates Ag processing and maturation of DCs and contributes to pulmonary inflammation and immune response against D. farinae. Furthermore, an additional yet to be identified resident cell type is essential for the development of pulmonary inflammation, likely a cell in which group V sPLA(2) is upregulated by D. farinae, and whose function is also regulated by group V sPLA(2).


Assuntos
Células Dendríticas/metabolismo , Hipersensibilidade/imunologia , Fosfolipases A2 Secretórias/imunologia , Pneumonia/imunologia , Pyroglyphidae/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Fosfolipases A2 do Grupo V/imunologia , Hipersensibilidade/complicações , Hipersensibilidade/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia/enzimologia , Pneumonia/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
EMBO Mol Med ; 2(5): 172-87, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20432503

RESUMO

Phospholipase A2 (PLA2) catalyses the release of arachidonic acid for generation of lipid mediators of inflammation and is crucial in diverse inflammatory processes. The functions of the secretory PLA2 enzymes (sPLA2), numbering nine members in humans, are poorly understood, though they have been shown to participate in lipid mediator generation and the associated inflammation. To further understand the roles of sPLA2 in disease, we quantified the expression of these enzymes in the synovial fluid in rheumatoid arthritis and used gene-deleted mice to examine their contribution in a mouse model of autoimmune erosive inflammatory arthritis. Contrary to expectation, we find that the group V sPLA2 isoform plays a novel anti-inflammatory role that opposes the pro-inflammatory activity of group IIA sPLA2. Mechanistically, group V sPLA2 counter-regulation includes promotion of immune complex clearance by regulating cysteinyl leukotriene synthesis. These observations identify a novel anti-inflammatory function for a PLA2 and identify group V sPLA2 as a potential biotherapeutic for treatment of immune-complex-mediated inflammation.


Assuntos
Anti-Inflamatórios/imunologia , Complexo Antígeno-Anticorpo/imunologia , Artrite Reumatoide/enzimologia , Artrite Reumatoide/imunologia , Fosfolipases A2 Secretórias/imunologia , Animais , Artrite Reumatoide/genética , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfolipases A2 Secretórias/genética , Líquido Sinovial/enzimologia , Líquido Sinovial/imunologia
18.
J Immunol ; 184(9): 5325-32, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20363972

RESUMO

We generated Fas-activated serine threonine phosphoprotein (FAST)-deficient mice (FAST(-/-)) to study the in vivo role of FAST in immune system function. In a model of house dust mite-induced allergic pulmonary inflammation, wild type mice develop a mixed cellular infiltrate composed of eosinophils, lymphocytes, and neutrophils. FAST(-/-) mice develop airway inflammation that is distinguished by the near absence of neutrophils. Similarly, LPS-induced alveolar neutrophil recruitment is markedly reduced in FAST(-/-) mice compared with wild type controls. This is accompanied by reduced concentrations of cytokines (TNF-alpha and IL-6 and -23) and chemoattractants (MIP-2 and keratinocyte chemoattractant) in bronchoalveolar lavage fluids. Because FAST(-/-) neutrophils exhibit normal chemotaxis and survival, impaired neutrophil recruitment is likely to be due to reduced production of chemoattractants within the pulmonary parenchyma. Studies using bone marrow chimeras implicate lung resident hematopoietic cells (e.g., pulmonary dendritic cells and/or alveolar macrophages) in this process. In conclusion, our results introduce FAST as a proinflammatory factor that modulates the function of lung resident hematopoietic cells to promote neutrophil recruitment and pulmonary inflammation.


Assuntos
Proteínas Mitocondriais/fisiologia , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/patologia , Receptor fas/fisiologia , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Alérgenos/administração & dosagem , Alérgenos/imunologia , Animais , Células Cultivadas , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Dermatophagoides pteronyssinus/imunologia , Poeira/imunologia , Feminino , Hematopoese/genética , Hematopoese/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas de Ligação a RNA/fisiologia , Hipersensibilidade Respiratória/genética , Antígeno-1 Intracelular de Células T
19.
Proc Natl Acad Sci U S A ; 106(28): 11685-90, 2009 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-19561298

RESUMO

The cysteinyl leukotrienes (cys-LTs) are proinflammatory lipid mediators acting on the type 1 cys-LT receptor (CysLT(1)R) to mediate smooth muscle constriction and vascular permeability. GPR17, a G protein-coupled orphan receptor with homology to the P2Y and cys-LT receptors, failed to mediate calcium flux in response to leukotriene (LT) D(4) with stable transfectants. However, in stable cotransfections of 6xHis-tagged GPR17 with Myc-tagged CysLT(1)R, the robust CysLT(1)R-mediated calcium response to LTD(4) was abolished. The membrane expression of the CysLT(1)R analyzed by FACS with anti-Myc Ab was not reduced by the cotransfection, yet both LTD(4)-elicited ERK phosphorylation and the specific binding of [(3)H]LTD(4) to microsomal membranes were fully inhibited. CysLT(1)R and GPR17 expressed in transfected cells were coimmunoprecipitated and identified by Western blots, and confocal immunofluorescence microscopy revealed that GPR17 and CysLT(1)R colocalize on the cell surface of human peripheral blood monocytes. Lentiviral knockdown of GPR17 in mouse bone marrow-derived macrophages (BMMPhis) increased both the membrane expression of CysLT(1)R protein by FACS analysis and the LTD(4)-elicited calcium flux in a dose-dependent manner as compared with control BMMPhis, indicating a negative regulatory function of GPR17 for CysLT(1)R in a primary cell. In IgE-dependent passive cutaneous anaphylaxis, GPR17-deficient mice showed a marked and significant increase in vascular permeability as compared with WT littermates, and this vascular leak was significantly blocked by pretreatment of the mice with the CysLT(1)R antagonist, MK-571. Taken together, our findings suggest that GPR17 is a ligand-independent, constitutive negative regulator for the CysLT(1)R that suppresses CysLT(1)R-mediated function at the cell membrane.


Assuntos
Permeabilidade Capilar/genética , Leucotrieno D4/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Leucotrienos/metabolismo , Animais , Western Blotting , Cálcio/metabolismo , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Imunoprecipitação , Camundongos , Microscopia de Fluorescência , Monócitos/metabolismo , Proteínas do Tecido Nervoso/genética , Receptores Acoplados a Proteínas G/genética
20.
J Immunol ; 182(8): 4891-8, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19342668

RESUMO

Phospholipase A(2) (PLA(2)) hydrolyzes the sn-2 position of cell membrane phospholipids to release fatty acids and lysophospholipids. We have previously reported that group V secretory PLA(2) (sPLA(2)) translocates from the Golgi and recycling endosomes of mouse peritoneal macrophages to newly formed phagosomes and regulates the phagocytosis of zymosan, suggesting a role in innate immunity. Here we report that in macrophages lacking group V sPLA(2), phagosome maturation was reduced 50-60% at early time points while the binding of zymosan was unimpaired. The ability of group V sPLA(2) to regulate phagocytosis extended to phagocytosis of IgG- and complement-opsonized sheep RBC. Moreover, macrophages lacking group V sPLA(2) had delays in phagocytosis, phagosome maturation, and killing of Candida albicans. Cytokine production and eicosanoid generation were not impaired by the lack of group V sPLA(2). Furthermore, in a model of systemic candidiasis, mice lacking group V sPLA(2) had an increased fungal burden in the kidney, liver, and spleen at day 7 postinfection and increased mortality. Thus, group V sPLA(2) regulates phagocytosis through major phagocytic receptors and contributes to the innate immune response against C. albicans by regulating phagocytosis and killing through a mechanism that is likely dependent on phagolysosome fusion.


Assuntos
Candida albicans/imunologia , Fosfolipases A2 do Grupo V/metabolismo , Imunidade Inata/imunologia , Fagossomos/enzimologia , Fagossomos/imunologia , Animais , Candidíase/genética , Candidíase/imunologia , Candidíase/metabolismo , Candidíase/patologia , Fosfolipases A2 do Grupo V/deficiência , Fosfolipases A2 do Grupo V/genética , Lectinas Tipo C , Macrófagos/enzimologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Fagocitose , Taxa de Sobrevida , Fator de Necrose Tumoral alfa/biossíntese , Zimosan/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...