Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 195(3): 1139-51, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26101324

RESUMO

Blood-derived monocytes remove apoptotic cells and terminate inflammation in settings as diverse as atherosclerosis and Alzheimer's disease. They express high levels of the proresolving receptor ALX/FPR2, which is activated by the protein annexin A1 (ANXA1), found in high abundance in inflammatory exudates. Using primary human blood monocytes from healthy donors, we identified ANXA1 as a potent CD14(+)CD16(-) monocyte chemoattractant, acting via ALX/FPR2. Downstream signaling pathway analysis revealed the p38 MAPK-mediated activation of a calcium independent phospholipase A2 with resultant synthesis of lysophosphatidic acid (LPA) driving chemotaxis through LPA receptor 2 and actin cytoskeletal mobilization. In vivo experiments confirmed ANXA1 as an independent phospholipase A2-dependent monocyte recruiter; congruently, monocyte recruitment was significantly impaired during ongoing zymosan-induced inflammation in AnxA1(-/-) or alx/fpr2/3(-/-) mice. Using a dorsal air-pouch model, passive transfer of apoptotic neutrophils between AnxA1(-/-) and wild-type mice identified effete neutrophils as the primary source of soluble ANXA1 in inflammatory resolution. Together, these data elucidate a novel proresolving network centered on ANXA1 and LPA generation and identify previously unappreciated determinants of ANXA1 and ALX/FPR2 signaling in monocytes.


Assuntos
Anexina A1/imunologia , Apoptose/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Receptores de Ácidos Lisofosfatídicos/imunologia , Citoesqueleto de Actina/metabolismo , Animais , Anexina A1/genética , Células Cultivadas , Ativação Enzimática/imunologia , Humanos , Inflamação/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Lisofosfolipídeos/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/transplante , Fosfolipases A2 Independentes de Cálcio/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores de Formil Peptídeo/biossíntese , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores de IgG/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Zimosan , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Proc Natl Acad Sci U S A ; 111(52): 18685-90, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25512512

RESUMO

Sepsis is characterized by overlapping phases of excessive inflammation temporally aligned with an immunosuppressed state, defining a complex clinical scenario that explains the lack of successful therapeutic options. Here we tested whether the formyl-peptide receptor 2/3 (Fpr2/3)--ortholog to human FPR2/ALX (receptor for lipoxin A4)--exerted regulatory and organ-protective functions in experimental sepsis. Coecal ligature and puncture was performed to obtain nonlethal polymicrobial sepsis, with animals receiving antibiotics and analgesics. Clinical symptoms, temperature, and heart function were monitored up to 24 h. Peritoneal lavage and plasma samples were analyzed for proinflammatory and proresolving markers of inflammation and organ dysfunction. Compared with wild-type mice, Fpr2/3(-/-) animals exhibited exacerbation of disease severity, including hypothermia and cardiac dysfunction. This scenario was paralleled by higher levels of cytokines [CXCL1 (CXC receptor ligand 1), CCL2 (CC receptor ligand 2), and TNFα] as quantified in cell-free biological fluids. Reduced monocyte recruitment in peritoneal lavages of Fpr2/3(-/-) animals was reflected by a higher granulocyte/monocyte ratio. Monitoring Fpr2/3(-/-) gene promoter activity with a GFP proxy marker revealed an over threefold increase in granulocyte and monocyte signals at 24 h post-coecal ligature and puncture, a response mediated by TNFα. Treatment with a receptor peptido-agonist conferred protection against myocardial dysfunction in wild-type, but not Fpr2/3(-/-), animals. Therefore, coordinated physio-pharmacological analyses indicate nonredundant modulatory functions for Fpr2/3 in experimental sepsis, opening new opportunities to manipulate the host response for therapeutic development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Granulócitos/metabolismo , Monócitos/metabolismo , Receptores de Formil Peptídeo/metabolismo , Sepse/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Modelos Animais de Doenças , Granulócitos/patologia , Humanos , Camundongos , Camundongos Knockout , Monócitos/patologia , Peritônio/metabolismo , Peritônio/patologia , Receptores de Formil Peptídeo/genética , Sepse/genética , Sepse/patologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
3.
Proc Natl Acad Sci U S A ; 110(45): 18232-7, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24108355

RESUMO

Formyl-peptide receptor type 2 (FPR2), also called ALX (the lipoxin A4 receptor), conveys the proresolving properties of lipoxin A4 and annexin A1 (AnxA1) and the proinflammatory signals elicited by serum amyloid protein A and cathelicidins, among others. We tested here the hypothesis that ALX might exist as homo- or heterodimer with FPR1 or FPR3 (the two other family members) and operate in a ligand-biased fashion. Coimmunoprecipitation and bioluminescence resonance energy transfer assays with transfected HEK293 cells revealed constitutive dimerization of the receptors; significantly, AnxA1, but not serum amyloid protein A, could activate ALX homodimers. A p38/MAPK-activated protein kinase/heat shock protein 27 signaling signature was unveiled after AnxA1 application, leading to generation of IL-10, as measured in vitro (in primary monocytes) and in vivo (after i.p. injection in the mouse). The latter response was absent in mice lacking the ALX ortholog. Using a similar approach, ALX/FPR1 heterodimerization evoked using the panagonist peptide Ac2-26, identified a JNK-mediated proapoptotic path that was confirmed in primary neutrophils. These findings provide a molecular mechanism that accounts for the dual nature of ALX and indicate that agonist binding and dimerization state contribute to the conformational landscape of FPRs.


Assuntos
Anexina A1/metabolismo , Conformação Proteica , Receptores de Formil Peptídeo/química , Receptores de Formil Peptídeo/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Técnicas de Transferência de Energia por Ressonância de Bioluminescência , Dimerização , Células HEK293 , Humanos , Imunoprecipitação , Interleucina-10/metabolismo , Camundongos , Dados de Sequência Molecular , Proteína Amiloide A Sérica/metabolismo
4.
J Biol Chem ; 289(52): 36166-78, 2014 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-25326384

RESUMO

Formyl-peptide receptor type 2 (FPR2; also called ALX because it is the receptor for lipoxin A4) sustains a variety of biological responses relevant to the development and control of inflammation, yet the cellular regulation of this G-protein-coupled receptor remains unexplored. Here we report that, in response to peptide agonist activation, FPR2/ALX undergoes ß-arrestin-mediated endocytosis followed by rapid recycling to the plasma membrane. We identify a transplantable recycling sequence that is both necessary and sufficient for efficient receptor recycling. Furthermore, removal of this C-terminal recycling sequence alters the endocytic fate of FPR2/ALX and evokes pro-apoptotic effects in response to agonist activation. This study demonstrates the importance of endocytic recycling in the anti-apoptotic properties of FPR2/ALX and identifies the molecular determinant required for modulation of this process fundamental for the control of inflammation.


Assuntos
Apoptose , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Sequência de Aminoácidos , Arrestinas/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptores de Formil Peptídeo/química , Receptores de Lipoxinas/química , beta-Arrestinas
5.
J Autoimmun ; 58: 1-11, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25619792

RESUMO

Annexin-A1 (Anx-A1) is an endogenous anti-inflammatory molecule and while described as a repressor of innate immune responses, the role of Anx-A1 in adaptive immunity, and in particular in T helper (Th) cell responses, remains controversial. We have used a T-cell mediated mouse model of retinal autoimmune disease to unravel the role of Anx-A1 in the development of autoreactive Th cell responses and pathology. RBP1-20-immunized C57BL/6 Anx-A1(-/-) mice exhibit significantly enhanced retinal inflammation and pathology as a result of an uncontrolled proliferation and activation of Th17 cells. This is associated with a limited capacity to induce SOCS3, resulting in un-restricted phosphorylation of STAT3. RBP1-20-specific CD4(+) cells from immunized Anx-A1(-/-) animals generated high levels of Th17 cells-associated cytokines. Following disease induction, daily systemic administration of human recombinant Anx-A1 (hrAnx-A1), during the afferent phase of disease, restrained autoreactive CD4(+) cell proliferation, reduced expression of pro-inflammatory cytokines IL-17, IFN-γ and IL-6 and attenuated autoimmune retinal inflammatory disease. Furthermore, in man, Anx-A1 serum levels when measured in active uveitis patient sera were low and associated with the detection of IgM and IgG anti-Anx-A1 antibodies when compared to healthy individuals. This data supports Anx-A1 as an early and critical regulator of Th17 cell driven autoimmune diseases such as uveitis.


Assuntos
Anexina A1/administração & dosagem , Doenças Autoimunes/imunologia , Proteínas Recombinantes/administração & dosagem , Células Th17/efeitos dos fármacos , Uveíte/imunologia , Animais , Anexina A1/genética , Doenças Autoimunes/induzido quimicamente , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Proteínas do Olho/imunologia , Humanos , Mediadores da Inflamação/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/imunologia , Proteínas Recombinantes/genética , Proteínas de Ligação ao Retinol/imunologia , Fator de Transcrição STAT3/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Células Th17/fisiologia , Uveíte/induzido quimicamente
6.
Blood ; 122(4): 608-17, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23733341

RESUMO

Endogenous protective pathways mitigate the overshooting of inflammation after sterile or infectious injury. Here we report that formyl peptide receptor 2 (Fpr2/3) null mice display a major phenotype with exacerbated vascular inflammation observed postischemia reperfusion (IR) injury of the mesenteric artery, characterized by marked neutrophil adhesion and extravasation as visualized by intravital microscopy. Analysis of endogenous agonists for Fpr2/3 revealed that lipoxin A4 (LXA4) was generated by platelet/neutrophil aggregates during ischemia: this cellular response was attenuated in Fpr2/3(-/-) mice; hence, LXA4 levels were lower after 30 minutes' ischemia, and associated with augmented vascular inflammation in the reperfusion (45-180 minutes) phase. Exogenous delivery of LXA4 attenuated IR-mediated inflammation in Fpr2/3(+/+) but not Fpr2/3(-/-) mice; conversely, an Fpr2/3 antagonist skewed the vascular phenotype of Fpr2/3(+/+) mice to that of Fpr2/3(-/-) animals. Such LXA4-based circuit could be activated by aspirin (30-100 mg/kg), which triggered formation of 15-epi-LXA4 in wild-type mice, yet it was effective in Fpr2/3(-/-) mice. In summary, we propose that during ischemia, neutrophil Fpr2/3 controls platelet/neutrophil aggregates with the rapid generation of circulating LXA4, which in turn modulates downstream vascular inflammatory responses evident during the reperfusion phase.


Assuntos
Aspirina/farmacologia , Vasos Sanguíneos , Citoproteção , Lipoxinas/farmacologia , Microcirculação/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Citoproteção/efeitos dos fármacos , Citoproteção/genética , Lipoxinas/química , Masculino , Camundongos , Camundongos Knockout , Microcirculação/genética , Modelos Biológicos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Receptores de Formil Peptídeo/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
7.
Nature ; 461(7268): 1287-91, 2009 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-19865173

RESUMO

A growing body of evidence indicates that resolution of acute inflammation is an active process. Resolvins are a new family of lipid mediators enzymatically generated within resolution networks that possess unique and specific functions to orchestrate catabasis, the phase in which disease declines. Resolvin D2 (RvD2) was originally identified in resolving exudates, yet its individual contribution in resolution remained to be elucidated. Here, we establish RvD2's potent stereoselective actions in reducing excessive neutrophil trafficking to inflammatory loci. RvD2 decreased leukocyte-endothelial interactions in vivo by endothelial-dependent nitric oxide production, and by direct modulation of leukocyte adhesion receptor expression. In mice with microbial sepsis initiated by caecal ligation and puncture, RvD2 sharply decreased both local and systemic bacterial burden, excessive cytokine production and neutrophil recruitment, while increasing peritoneal mononuclear cells and macrophage phagocytosis. These multi-level pro-resolving actions of RvD2 translate to increased survival from sepsis induced by caecal ligation and puncture and surgery. Together, these results identify RvD2 as a potent endogenous regulator of excessive inflammatory responses that acts via multiple cellular targets to stimulate resolution and preserve immune vigilance.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Sepse/imunologia , Sepse/microbiologia , Animais , Ácidos Docosa-Hexaenoicos/síntese química , Ácidos Docosa-Hexaenoicos/química , Células Endoteliais/metabolismo , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/isolamento & purificação , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Cavidade Peritoneal/citologia , Cavidade Peritoneal/microbiologia , Peritonite/imunologia , Peritonite/metabolismo , Peritonite/microbiologia , Fagocitose , Espécies Reativas de Oxigênio/metabolismo , Sepse/metabolismo
8.
J Pharmacol Exp Ther ; 351(1): 96-104, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25077524

RESUMO

Hydrogen sulfide (H2S) is a gaseous mediator synthesized in mammalian tissues by three main enzymes-cystathionine-ß-synthase (CBS), cystathionine-γ-lyase (CSE), and 3-mercaptopyruvate-sulfurtransferase-and its levels increase under inflammatory conditions or sepsis. Since H2S and H2S-releasing molecules afford inhibitory properties in leukocyte trafficking, we tested whether endogenous annexin A1 (AnxA1), a glucocorticoid-regulated inhibitor of inflammation acting through formylated-peptide receptor 2 (ALX), could display intermediary functions in the anti-inflammatory profile of H2S. We first investigated whether endogenous AnxA1 could modulate H2S biosynthesis. To this end, a marked increase in CBS and/or CSE gene products was quantified by quantitative real-time polymerase chain reaction in aortas, kidneys, and spleens collected from AnxA1(-/-) mice, as compared with wild-type animals. When lipopolysaccharide-stimulated bone marrow-derived macrophages were studied, H2S-donor sodium hydrosulfide (NaHS) counteracted the increased expression of inducible nitric oxide synthase and cyclooxygenase 2 mRNA evoked by the endotoxin, yet it was inactive in macrophages harvested from AnxA1(-/-) mice. Next we studied the effect of in vivo administration of NaHS in a model of interleukin-1ß (IL-1ß)-induced mesenteric inflammation. AnxA1(+/+) mice treated with NaHS (100 µmol/kg) displayed inhibition of IL-1ß-induced leukocyte adhesion/emigration in the inflamed microcirculation, not observed in AnxA1(-/-) animals. These results were translated by testing human neutrophils, where NaHS (10-100 µM) prompted an intense mobilization (>50%) of AnxA1 from cytosol to cell surface, an event associated with inhibition of cell/endothelium interaction under flow. Taken together, these data strongly indicate the existence of a positive interlink between AnxA1 and H2S pathway, with nonredundant functions in the control of experimental inflammation.


Assuntos
Anexina A1/metabolismo , Sulfeto de Hidrogênio/metabolismo , Animais , Anexina A1/genética , Aorta/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Adesão Celular , Movimento Celular , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Interleucina-1beta/farmacologia , Rim/metabolismo , Leucócitos/metabolismo , Leucócitos/fisiologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Baço/metabolismo , Sulfetos/farmacologia
9.
FASEB J ; 27(1): 368-78, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23038751

RESUMO

Obesity-related metabolic disorders are characterized by mild chronic inflammation, leukocyte infiltration, and tissue fibrosis as a result of adipocytokine production from the expanding white adipose tissue. Annexin A1 (AnxA1) is an endogenous glucocorticoid regulated protein, which modulates systemic anti-inflammatory processes and, therefore, may be altered with increasing adiposity in humans. Paradoxically, we found that plasma AnxA1 concentrations inversely correlated with BMI, total percentage body fat, and waist-to-hip ratio in human subjects. Plasma AnxA1 was also inversely correlated with plasma concentrations of the acute-phase protein, C-reactive protein (CRP), and the adipocytokine leptin, suggesting that as systemic inflammation increases, anti-inflammatory AnxA1 is reduced. In addition, AnxA1 gene expression and protein were significantly up-regulated during adipogenesis in a human adipocyte cell line compared to vehicle alone, demonstrating for the first time that AnxA1 is expressed and excreted from human adipocytes. These data demonstrate a failure in the endogenous anti-inflammatory system to respond to increasing systemic inflammation resulting from expanding adipose tissue, a condition strongly linked to the development of type 2 diabetes and cardiovascular disease. These data raise the possibility that a reduction in plasma AnxA1 may contribute to the chronic inflammatory phenotype observed in human obesity.


Assuntos
Anexina A1/sangue , Obesidade/sangue , Adulto , Biomarcadores/sangue , Proteína C-Reativa/metabolismo , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Mediadores da Inflamação/sangue , Leptina/sangue , Masculino , Reação em Cadeia da Polimerase em Tempo Real
10.
J Biol Chem ; 287(29): 24690-7, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22610094

RESUMO

Understanding how proresolving agonists selectively activate FPR2/ALX is a crucial step in the clarification of proresolution molecular networks that can be harnessed for the design of novel therapeutics for inflammatory disease. FPR2/ALX, a G protein-coupled receptor belonging to the formyl peptide receptor (FPR) family, conveys the biological functions of a variety of ligands, including the proresolution mediators annexin A1 (AnxA1) and lipoxin A(4), as well as the activating and proinflammatory protein serum amyloid A. FPR2/ALX is the focus of intense screening for novel anti-inflammatory therapeutics, and the small molecule compound 43 was identified as a receptor ligand. Here, we used chimeric FPR1 and FPR2/ALX clones (stably transfected in HEK293 cells) to identify the N-terminal region and extracellular loop II as the FPR2/ALX domain required for AnxA1-mediated signaling. Genomic responses were also assessed with domain-specific effects emerging, so the N-terminal region is required for AnxA1 induction of JAG1 and JAM3, whereas it is dispensable for modulation of SGPP2. By comparison, serum amyloid A non-genomic responses were reliant on extracellular loops I and II, whereas the small molecule compound 43 activated extracellular loop I with downstream signaling dependent on transmembrane region II. In desensitization experiments, the N-terminal region was dispensable for AnxA1-induced FPR2/ALX down-regulation in both the homologous and heterologous desensitization modes.


Assuntos
Anexina A1/química , Anexina A1/metabolismo , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Western Blotting , Cálcio/metabolismo , Citometria de Fluxo , Células HEK293 , Humanos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Receptores de Formil Peptídeo/química , Receptores de Lipoxinas/química , Proteína Amiloide A Sérica/farmacologia , Transdução de Sinais/efeitos dos fármacos
11.
Arterioscler Thromb Vasc Biol ; 32(8): 1970-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22499990

RESUMO

OBJECTIVE: Resolvin D1 (RvD1) limits neutrophil recruitment during acute inflammation and is derived from omega-3 docosahexaenoic acid to promote catabasis. The contribution of its specific receptors, the lipoxin A(4)/Annexin-A1 receptor formyl-peptide receptor 2 (FPR2/ALX) and the orphan receptor G-protein-coupled receptor 32 (GPR32) are of considerable interest. METHODS AND RESULTS: RvD1 reduced human polymorphonuclear leukocytes recruitment to endothelial cells under shear conditions as quantified using a flow chamber system. Receptor-specific antibodies blocked these anti-inflammatory actions of RvD1, with low (1 nmol/L) concentrations sensitive to GPR32 blockade, while the higher (10 nmol/L) concentration appeared FPR2/ALX-specific. Interestingly, polymorphonuclear leukocytes surface expression of FPR2/ALX but not GPR32 increased following activation with pro-inflammatory stimuli, corresponding with secretory vesicle mobilization. Lipid mediator metabololipidomics carried out with 24-hour exudates revealed that RvD1 in vivo gave a significant reduction in the levels of a number of pro-inflammatory mediators including prostaglandins and leukotriene B(4). These actions of RvD1 were abolished in fpr2 null mice. CONCLUSIONS: Pro-resolving lipid mediators and their receptors, such as RvD1 and the 2 G-protein-coupled receptors, studied here regulate resolution and may provide new therapeutic strategies for diseases with a vascular inflammatory component.


Assuntos
Ácidos Docosa-Hexaenoicos/fisiologia , Inflamação/patologia , Neutrófilos/fisiologia , Animais , Movimento Celular , Humanos , Masculino , Camundongos , Receptores de Formil Peptídeo/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Lipoxinas/fisiologia
12.
J Immunol ; 186(8): 4905-14, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21398608

RESUMO

The importance of proresolving mediators in the overall context of the resolution of acute inflammation is well recognized, although little is known about whether these anti-inflammatory and proresolving molecules act in concert. In this article, we focused on lipoxin A(4) (LXA(4)) and annexin A1 (AnxA1) because these two very different mediators converge on a single receptor, formyl peptide receptor type 2 (FPR2/ALX). Addition of LXA(4) to human polymorphonuclear leukocytes (PMNs) provoked a concentration- and time-dependent mobilization of AnxA1 onto the plasma membrane, as determined by Western blotting and flow cytometry analyses. This property was shared by another FPR2/ALX agonist, antiflammin-2, and partly by fMLF or peptide Ac2-26 (an AnxA1 derivative that can activate all three members of the human FPR family). An FPR2/ALX antagonist blocked AnxA1 mobilization activated by LXA(4) and antiflammin-2. Analysis of PMN degranulation patterns and phospho-AnxA1 status suggested a model in which the two FPR2/ALX agonists mobilize the cytosolic (and not the granular) pool of AnxA1 through an intermediate phosphorylation step. Intravital microscopy investigations of the inflamed mesenteric microvasculature of wild-type and AnxA1(-/-) mice revealed that LXA(4) provoked leukocyte detachment from the postcapillary venule endothelium in the former (>50% within 10 min; p < 0.05), but not the latter genotype (∼15%; NS). Furthermore, recruitment of Gr1(+) cells into dorsal air-pouches, inflamed with IL-1ß, was significantly attenuated by LXA(4) in wild-type, but not AnxA1(-/-), mice. Collectively, these data prompt us to propose the existence of an endogenous network in anti-inflammation centered on PMN AnxA1 and activated by selective FPR2/ALX agonists.


Assuntos
Microvasos/imunologia , Neutrófilos/imunologia , Receptores de Formil Peptídeo/imunologia , Receptores de Lipoxinas/imunologia , Animais , Anexina A1/genética , Anexina A1/imunologia , Anexina A1/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Western Blotting , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/prevenção & controle , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Lipoxinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvasos/metabolismo , Microvasos/patologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Transporte Proteico/efeitos dos fármacos , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Fatores de Tempo
13.
J Immunol ; 186(10): 5543-7, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21460209

RESUMO

Endogenous microparticles (MPs) were systematically profiled during the time course of self-limited inflammation. Precursors for specialized proresolving lipid mediators were identified in MPs from inflammatory exudates using liquid chromatography tandem mass spectrometry-based metabolomics. Hence, we postulated that formation of anti-inflammatory and proresolving lipid mediators could underlie beneficial effects attributed to MPs and that this process could serve as a basis for biomimicry. Using human neutrophil-derived MPs, we constructed novel nanoparticles (NPs) containing aspirin-triggered resolvin D1 or a lipoxin A(4) analog. Enriched NPs dramatically reduced polymorphonuclear cell influx in murine peritonitis, shortened resolution intervals, and exhibited proresolving actions accelerating keratinocyte healing. The enriched NPs protected against inflammation in the temporomandibular joint. These findings indicate that humanized NPs, termed nano-proresolving medicines, are mimetics of endogenous resolving mechanisms, possess potent beneficial bioactions, can reduce nanotoxicity, and offer new therapeutic approaches.


Assuntos
Anti-Inflamatórios , Ácidos Docosa-Hexaenoicos , Inflamação/tratamento farmacológico , Lipoxinas/farmacologia , Nanopartículas/uso terapêutico , Transtornos da Articulação Temporomandibular/tratamento farmacológico , Cicatrização/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Aspirina , Movimento Celular/efeitos dos fármacos , Cromatografia Líquida , Ácidos Docosa-Hexaenoicos/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Ácidos Docosa-Hexaenoicos/uso terapêutico , Humanos , Inflamação/prevenção & controle , Queratinócitos/efeitos dos fármacos , Lipídeos/farmacologia , Lipoxinas/metabolismo , Lipoxinas/uso terapêutico , Masculino , Camundongos , Microesferas , Mimetismo Molecular , Neutrófilos/imunologia , Neutrófilos/metabolismo , Peritonite/tratamento farmacológico , Peritonite/imunologia , Espectrometria de Massas em Tandem , Articulação Temporomandibular/efeitos dos fármacos , Articulação Temporomandibular/imunologia , Cicatrização/imunologia
14.
J Immunol ; 184(5): 2611-2619, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20107188

RESUMO

The human formyl-peptide receptor (FPR)-2 is a G protein-coupled receptor that transduces signals from lipoxin A(4), annexin A1, and serum amyloid A (SAA) to regulate inflammation. In this study, we report the creation of a novel mouse colony in which the murine FprL1 FPR2 homologue, Fpr2, has been deleted and describe its use to explore the biology of this receptor. Deletion of murine fpr2 was verified by Southern blot analysis and PCR, and the functional absence of the G protein-coupled receptor was confirmed by radioligand binding assays. In vitro, Fpr2(-/-) macrophages had a diminished response to formyl-Met-Leu-Phe itself and did not respond to SAA-induced chemotaxis. ERK phosphorylation triggered by SAA was unchanged, but that induced by the annexin A1-derived peptide Ac2-26 or other Fpr2 ligands, such as W-peptide and compound 43, was attenuated markedly. In vivo, the antimigratory properties of compound 43, lipoxin A(4), annexin A1, and dexamethasone were reduced notably in Fpr2(-/-) mice compared with those in wild-type littermates. In contrast, SAA stimulated neutrophil recruitment, but the promigratory effect was lost following Fpr2 deletion. Inflammation was more marked in Fpr2(-/-) mice, with a pronounced increase in cell adherence and emigration in the mesenteric microcirculation after an ischemia-reperfusion insult and an augmented acute response to carrageenan-induced paw edema, compared with that in wild-type controls. Finally, Fpr2(-/-) mice exhibited higher sensitivity to arthrogenic serum and were completely unable to resolve this chronic pathology. We conclude that Fpr2 is an anti-inflammatory receptor that serves varied regulatory functions during the host defense response. These data support the development of Fpr2 agonists as novel anti-inflammatory therapeutics.


Assuntos
Inflamação/metabolismo , Leucócitos/metabolismo , Ligantes , Receptores de Formil Peptídeo/fisiologia , Sequência de Aminoácidos , Animais , Anexina A1/metabolismo , Anexina A1/farmacologia , Carragenina , Quimiotaxia/efeitos dos fármacos , Edema/induzido quimicamente , Edema/genética , Edema/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Citometria de Fluxo , Immunoblotting , Inflamação/induzido quimicamente , Inflamação/genética , Leucócitos/citologia , Lipoxinas/metabolismo , Lipoxinas/farmacologia , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Dados de Sequência Molecular , N-Formilmetionina Leucil-Fenilalanina/análogos & derivados , N-Formilmetionina Leucil-Fenilalanina/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Peptídeos/metabolismo , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Zimosan
15.
J Exp Med ; 201(5): 671-4, 2005 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-15753201

RESUMO

The inflammatory response protects the body against infection and injury but can itself become deregulated with deleterious consequences to the host. It is now clear that several endogenous biochemical pathways activated during defense reactions can counterregulate inflammation. New experimental evidence adds resolvin E1 to this group of endogenous inhibitors and provides further rationale for the beneficial effects of dietary supplementation with fish oils. It also highlights an unexpected twist in the pharmacology of aspirin.


Assuntos
Anti-Inflamatórios/farmacologia , Gorduras na Dieta/farmacologia , Inflamação/dietoterapia , Inflamação/tratamento farmacológico , Animais , Aspirina/farmacologia , Ácidos Graxos Insaturados/farmacologia , Humanos , Inflamação/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
16.
BMC Immunol ; 12: 59, 2011 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22011168

RESUMO

BACKGROUND: The balancing functions of pro/anti-inflammatory mediators of the complex innate responses have been investigated in a variety of experimental inflammatory settings. Annexin-A1 (AnxA1) is one mediator of endogenous anti-inflammation, affording regulation of leukocyte trafficking and activation in many contexts, yet its role in lung pathologies has been scarcely investigated, despite being highly expressed in lung cells. Here we have applied the bleomycin lung fibrosis model to AnxA1 null mice over a 21-day time-course, to monitor potential impact of this mediator on the control of the inflammatory and fibrotic phases. RESULTS: Analyses in wild-type mice revealed strict spatial and temporal regulation of the Anxa1 gene, e.g. up-regulation in epithelial cells and infiltrated granulocytes at day 7, followed by augmented protein levels in alveolar macrophages by day 21. Absence of AnxA1 caused increases in: i) the degree of inflammation at day 7; and ii) indexes of fibrosis (assessed by deposition of hydroxyproline in the lung) at day 7 and 21. These alterations in AnxA1 null mice were paralleled by augmented TGF-ß1, IFN-γ and TNF-α generation compared to wild-type mice. Finally, treatment of wild type animals with an AnxA1 peptido-mimetic, given prophylactically (from day 0 to 21) or therapeutically (from day 14 onward), ameliorated both signs of inflammation and fibrosis. CONCLUSION: Collectively these data reveal a pathophysiological relevance for endogenous AnxA1 in lung inflammation and, more importantly, fibrosis, and may open new insights for the pharmacological treatment of lung fibrosis.


Assuntos
Células Epiteliais/metabolismo , Pulmão/metabolismo , Macrófagos Alveolares/metabolismo , Pneumonia/metabolismo , Fibrose Pulmonar/metabolismo , Animais , Anexina A1/genética , Anexina A1/metabolismo , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/metabolismo , Bleomicina/administração & dosagem , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Regulação da Expressão Gênica/genética , Hidroxiprolina/metabolismo , Mediadores da Inflamação/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/tratamento farmacológico , Pneumonia/genética , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/genética , Fibrose Pulmonar/imunologia , Fatores de Tempo
17.
Drug Metab Dispos ; 39(9): 1689-95, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21628499

RESUMO

In recent years, there has been increasing interest in hypothermia induced by paracetamol for therapeutic purposes, which, in some instances, has been reported as a side effect. Understanding the mechanism by which paracetamol induces hypothermia is therefore an important question. In this study, we investigated whether the novel metabolite of paracetamol, N-(4-hydroxyphenyl)arachidonylamide (AM404), which activates the cannabinoid (CB) and transient receptor potential vanilloid-1 (TRPV1) systems, mediates the paracetamol-induced hypothermia. The hypothermic response to 300 mg/kg paracetamol in CB(1) receptor (CB(1)R) and TRPV1 knockout mice was compared to wild-type mice. Hypothermia induced by paracetamol was also investigated in animals pretreated with the CB(1)R or TRPV1 antagonist 1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperdinyl-1H-pyrazole-3-carboxamide trifluoroacetate salt (AM251) or 4'-chloro-3-methoxycinnamanilide (SB366791), respectively. In CB(1)R or TRPV1 knockout mice, paracetamol induced hypothermia to the same extent as in wild-type mice. In addition, in C57BL/6 mice pretreated with AM251 or SB366791, paracetamol induced hypothermia to the same extent as in control mice. AM404 failed to induce hypothermia at pharmacological doses. Inhibition of fatty acid amide hydrolase (FAAH), which is involved in the metabolism of paracetamol to AM404, did not prevent the development of hypothermia with paracetamol. Paracetamol also induced hypothermia in FAAH knockout mice to the same extent as wild-type mice. We conclude that paracetamol induces hypothermia independent of cannabinoids and TRPV1 and that AM404 does not mediate this response. In addition, potential therapeutic value of combinational drug-induced hypothermia is supported by experimental evidence.


Assuntos
Acetaminofen/farmacologia , Ácidos Araquidônicos/farmacologia , Canabinoides/metabolismo , Hipotermia Induzida/métodos , Canais de Cátion TRPV/metabolismo , Amidoidrolases/metabolismo , Anilidas/farmacologia , Animais , Cinamatos/farmacologia , Hipotermia/induzido quimicamente , Hipotermia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética
18.
Arterioscler Thromb Vasc Biol ; 30(9): 1718-24, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20558817

RESUMO

OBJECTIVE: To determine whether the inhibitory action of the antiallergic cromone "mast cell stabilizing" drugs on polymorphonuclear leukocyte (PMN) trafficking is mediated through an annexin-A1 (Anx-A1) dependent mechanism. METHODS AND RESULTS: Intravital microscopy was used to monitor the actions of cromones in the inflamed microcirculation. Reperfusion injury provoked a dramatic increase in adherent and emigrated leukocytes in the mesenteric vascular bed, associated with augmented tissue levels of myeloperoxidase. Nedocromil, 2 to 20 mg/kg, significantly (P<0.05) inhibited cell adhesion and emigration, as well as myeloperoxidase release, in wild-type but not Anx-A1(-/-) mice. Short pretreatment of human PMNs with nedocromil, 10 nmol/L, inhibited cell adhesion (P<0.05) in the flow chamber assay, and this effect was reversed by specific anti-AnxA1 or a combination of antiformyl peptide receptors 1 and 2, but not irrelevant control, antibodies. Western blotting experiments revealed that cromones stimulate protein kinase C-dependent phosphorylation and release Anx-A1 in human PMNs. CONCLUSIONS: We propose a novel mechanism to explain the antiinflammatory actions of cromones on PMN trafficking, an effect that has long puzzled investigators.


Assuntos
Anexina A1/metabolismo , Antialérgicos/farmacologia , Anti-Inflamatórios/farmacologia , Adesão Celular/efeitos dos fármacos , Cromolina Sódica/farmacologia , Células Endoteliais/efeitos dos fármacos , Nedocromil/farmacologia , Neutrófilos/efeitos dos fármacos , Animais , Anexina A1/deficiência , Anexina A1/genética , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Masculino , Oclusão Vascular Mesentérica/tratamento farmacológico , Oclusão Vascular Mesentérica/imunologia , Oclusão Vascular Mesentérica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação/efeitos dos fármacos , Microscopia de Vídeo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Peritonite/tratamento farmacológico , Peritonite/imunologia , Peritonite/metabolismo , Peroxidase/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Transporte Proteico , Receptores de Formil Peptídeo/efeitos dos fármacos , Receptores de Formil Peptídeo/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/metabolismo , Fatores de Tempo
19.
Am J Pathol ; 174(1): 177-83, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19095957

RESUMO

The purpose of this study was twofold: to reveal cellular events associated with the protective role of endogenous annexin A1 (AnxA1) in inflammation and to highlight the potential involvement of members of the formyl peptide receptor (Fpr) family in this process. We found that wild-type, AnxA1-null, and Fpr1-null mice all displayed an intense neutrophil recruitment into the peritoneal cavity as assessed 4 hours after carrageenin injection, and that this recruitment was most pronounced in AnxA1-null mice. In addition, this cell influx could be inhibited by the AnxA1 pharmacophore peptide, Ac2-26, in wild-type, AnxA1-null, and Fpr1-null mice, but was restored when co-treated with the pan-receptor antagonist Boc2. Using the LacZ gene reporter assay, an enhancement of AnxA1 gene promoter activity in extravasated neutrophils was evident in AnxA1-null mice; again this response was reduced after peptide treatment. The lack of functional involvement of Fpr1 prompted us to monitor the structurally related receptor Fpr2. We report, for the first time, the ultrastructural immunocytochemical co-localization of Fpr2 with AnxA1 in neutrophils that migrate into the mesenteric microcirculation and extravasate into the peritoneal fluid. Collectively, these data provide in vivo support to the hypothesis that endogenous AnxA1 is an essential effector of endogenous anti-inflammation and provide an ultrastructural indication that this mediator interacts with Fpr2 in murine neutrophils. We believe that these findings could significantly affect the development of novel therapeutics, which are modeled after the anti-migratory actions of AnxA1.


Assuntos
Anexina A1/metabolismo , Quimiotaxia de Leucócito/imunologia , Inflamação/metabolismo , Neutrófilos/metabolismo , Animais , Anexina A1/ultraestrutura , Expressão Gênica , Imuno-Histoquímica , Inflamação/imunologia , Masculino , Camundongos , Camundongos Mutantes , Neutrófilos/imunologia , Neutrófilos/ultraestrutura , Receptores de Formil Peptídeo/imunologia , Receptores de Formil Peptídeo/metabolismo , Receptores de Formil Peptídeo/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
FASEB J ; 23(4): 985-96, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19029200

RESUMO

Dendritic cells play a key role in the adaptive immune system by influencing T-cell differentiation. Annexin-1 (Anx-A1) has recently been shown to modulate the adaptive immune response by regulating T-cell activation and differentiation. Here we investigated the role of endogenous Anx-A1 in dendritic cells as major cellular counterpart of T-cell-driven immune response. We found that Anx-A1(-/-) bone marrow-derived dendritic cells show an increased number of CD11c(+) cells expressing high levels of some maturation markers, such as CD40, CD54, and CD80, coupled to a decreased capacity to take up antigen compared to control Anx-A1(+/+) cells. However, analysis of LPS-treated dendritic cells from Anx-A1(-/-) mice demonstrated a diminished up-regulation of maturation markers, a decreased migratory activity in vivo, and an attenuated production of the inflammatory cytokines interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha, and IL-12. This defect was also accompanied by impaired nuclear factor (NF)-kappaB/DNA-binding activity and lack of Anx-A1 signaling, as demonstrated by the reduced activation of extracellular-signal regulated kinase (ERK)1/2 and Akt compared to cells from control littermates. As a consequence of this phenotype, Anx-A1(-/-) dendritic cells showed an impaired capacity to stimulate T-cell proliferation and differentiation in mixed leukocyte reaction. Together, these findings suggest that inhibition of Anx-A1 expression or function in dendritic cells might represent a useful way to modulate the adaptive immune response and pathogen-induced T-cell-driven immune diseases.


Assuntos
Anexina A1/análise , Células Dendríticas/citologia , Animais , Anexina A1/metabolismo , Células Apresentadoras de Antígenos/imunologia , Antígeno B7-1/imunologia , Antígeno B7-1/metabolismo , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Antígeno CD11c/imunologia , Antígeno CD11c/metabolismo , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/biossíntese , Células Dendríticas/fisiologia , Dextranos/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Corantes Fluorescentes/metabolismo , Lipopolissacarídeos/farmacologia , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/química , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA