Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Pharmacol Sci ; 148(1): 56-64, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34924130

RESUMO

Acute kidney injury (AKI) is a serious complication in critically ill patients. Accumulating evidences indicated that macrophages play an important pro-inflammatory role in AKI and isoliquiritigenin (ISL) can inhibit macrophagic inflammation, but its role in AKI and the underlying mechanism are unknown. The present study aims to investigate the renoprotective effect of ISL on AKI and the role of Formyl peptide receptors 2 (FPR2) in this process. In this study, cisplatin-induced AKI model and lipopolysaccharide-induced macrophage inflammatory model were employed to perform the in vivo and in vitro experiments. The results showed that ISL strongly relieved kidney injury and inhibited renal inflammation in vivo and suppress macrophagic inflammatory response in vitro. Importantly, it was found that FPR2 was significantly upregulated compared to the control group in AKI and LPS-induced macrophage, whereas it was strongly suppressed by ISL. Interestingly, overexpression of FPR2 with transfection of pcDNA3.1-FPR2 effectively reversed the anti-inflammatory effect of ISL in macrophage, suggesting that FPR2 may be the potential target for ISL to prevent inflammation and improve kidney injury of AKI. Take together, these findings indicated that ISL improved cisplantin-induced kidney injury by inhibiting FPR2 involved macrophagic inflammation, which may provide a potential therapeutic option for AKI.


Assuntos
Injúria Renal Aguda/genética , Injúria Renal Aguda/prevenção & controle , Chalconas/farmacologia , Chalconas/uso terapêutico , Cisplatino/efeitos adversos , Macrófagos/metabolismo , Receptores de Formil Peptídeo/antagonistas & inibidores , Receptores de Lipoxinas/antagonistas & inibidores , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Animais , Células Cultivadas , Chalconas/isolamento & purificação , Expressão Gênica/efeitos dos fármacos , Glycyrrhiza/química , Inflamação , Masculino , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Fitoterapia , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores de Formil Peptídeo/fisiologia , Receptores de Lipoxinas/genética , Receptores de Lipoxinas/metabolismo , Receptores de Lipoxinas/fisiologia , Regulação para Cima/efeitos dos fármacos
2.
J Med Chem ; 64(13): 9193-9216, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34138563

RESUMO

Failure to resolve inflammation underlies many prevalent pathologies. Recent insights have identified lipid mediators, typified by lipoxins (LXs), as drivers of inflammation resolution, suggesting potential therapeutic benefit. We report the asymmetric preparation of novel quinoxaline-containing synthetic-LXA4-mimetics (QNX-sLXms). Eight novel compounds were screened for their impact on inflammatory responses. Structure-activity relationship (SAR) studies showed that (R)-6 (also referred to as AT-02-CT) was the most efficacious and potent anti-inflammatory compound of those tested. (R)-6 significantly attenuated lipopolysaccharide (LPS)- and tumor-necrosis-factor-α (TNF-α)-induced NF-κB activity in monocytes and vascular smooth muscle cells. The molecular target of (R)-6 was investigated. (R)-6 activated the endogenous LX receptor formyl peptide receptor 2 (ALX/FPR2). The anti-inflammatory properties of (R)-6 were further investigated in vivo in murine models of acute inflammation. Consistent with in vitro observations, (R)-6 attenuated inflammatory responses. These results support the therapeutic potential of the lead QNX-sLXm (R)-6 in the context of novel inflammatory regulators.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Quinoxalinas/farmacologia , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Animais , Anti-Inflamatórios não Esteroides/síntese química , Anti-Inflamatórios não Esteroides/química , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Camundongos , Estrutura Molecular , Monócitos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Quinoxalinas/síntese química , Quinoxalinas/química , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
3.
J Med Chem ; 63(17): 9003-9019, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32407089

RESUMO

Formyl peptide receptor 2 (FPR2) agonists can stimulate resolution of inflammation and may have utility for treatment of diseases caused by chronic inflammation, including heart failure. We report the discovery of a potent and selective FPR2 agonist and its evaluation in a mouse heart failure model. A simple linear urea with moderate agonist activity served as the starting point for optimization. Introduction of a pyrrolidinone core accessed a rigid conformation that produced potent FPR2 and FPR1 agonists. Optimization of lactam substituents led to the discovery of the FPR2 selective agonist 13c, BMS-986235/LAR-1219. In cellular assays 13c inhibited neutrophil chemotaxis and stimulated macrophage phagocytosis, key end points to promote resolution of inflammation. Cardiac structure and functional improvements were observed in a mouse heart failure model following treatment with BMS-986235/LAR-1219.


Assuntos
Pirrolidinonas/química , Receptores de Formil Peptídeo/agonistas , Receptores de Lipoxinas/agonistas , Animais , Quimiotaxia/efeitos dos fármacos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/prevenção & controle , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Microssomos Hepáticos/metabolismo , Neutrófilos/citologia , Neutrófilos/fisiologia , Fagocitose/efeitos dos fármacos , Pirrolidinonas/metabolismo , Pirrolidinonas/farmacologia , Pirrolidinonas/uso terapêutico , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/genética , Receptores de Lipoxinas/metabolismo , Relação Estrutura-Atividade
4.
JCI Insight ; 3(6)2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29563331

RESUMO

While the treatment of inflammatory disorders is generally based on inhibiting factors that drive onset of inflammation, these therapies can compromise healing (NSAIDs) or dampen immunity against infections (biologics). In search of new antiinflammatories, efforts have focused on harnessing endogenous pathways that drive resolution of inflammation for therapeutic gain. Identification of specialized pro-resolving mediators (SPMs) (lipoxins, resolvins, protectins, maresins) as effector molecules of resolution has shown promise in this regard. However, their action on inflammatory resolution in humans is unknown. Here, we demonstrate using a model of UV-killed Escherichia coli-triggered skin inflammation that SPMs are biosynthesized at the local site at the start of resolution, coinciding with the expression of receptors that transduce their actions. These include receptors for lipoxin A4 (ALX/FPR2), resolvin E1 (ChemR23), resolvin D2 (GPR18), and resolvin D1 (GPR32) that were differentially expressed on the endothelium and infiltrating leukocytes. Administering SPMs into the inflamed site 4 hours after bacterial injection caused a reduction in PMN numbers over the ensuing 6 hours, the phase of active resolution in this model. These results indicate that in humans, the appearance of SPMs and their receptors is associated with the beginning of inflammatory resolution and that their therapeutic supplementation enhanced the resolution response.


Assuntos
Anti-Inflamatórios/farmacologia , Escherichia coli/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Pele/imunologia , Pele/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adolescente , Adulto , Vesícula/imunologia , Vesícula/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Eicosanoides/imunologia , Eicosanoides/farmacologia , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/farmacologia , Escherichia coli/efeitos da radiação , Humanos , Inflamação/tratamento farmacológico , Leucócitos/imunologia , Leucócitos/metabolismo , Lipoxinas/farmacologia , Masculino , Pessoa de Meia-Idade , Neutrófilos/efeitos dos fármacos , Receptores de Quimiocinas/metabolismo , Receptores de Formil Peptídeo/metabolismo , Receptores Acoplados a Proteínas G , Receptores de Lipoxinas/metabolismo , Pele/efeitos dos fármacos , Pele/patologia , Voluntários , Adulto Jovem
5.
FASEB J ; 31(8): 3393-3402, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28442547

RESUMO

Recent evidence suggests that specialized proresolving lipid mediators (SPMs) generated from docosahexaenoic acid (DHA) can modulate the vascular injury response. However, cellular sources for these autacoids within the vessel wall remain unclear. Here, we investigated whether isolated vascular cells and tissues can produce SPMs and assessed expression and subcellular localization of the key SPM biosynthetic enzyme 5-lipoxygenase (LOX) in vascular cells. Intact human arteries incubated with DHA ex vivo produced 17-hydroxy DHA (17-HDHA) and D-series resolvins, as assessed by liquid chromatography-tandem mass spectrometry. Addition of 17-HDHA to human arteries similarly increased resolvin production. Primary cultures of human saphenous vein endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) converted 17-HDHA to SPMs, including resolvin D1 (RvD1) and other D-series resolvins and protectins. This was accompanied by a rapid translocation of 5-LOX from nucleus to cytoplasm in both ECs and VSMCs, potentially facilitating SPM biosynthesis. Conditioned medium from cells exposed to 17-HDHA inhibited monocyte adhesion to TNF-α-stimulated EC monolayers. These downstream effects were partially reversed by antibodies against the RvD1 receptors ALX/FPR2 and GPR32. These results suggest that autocrine and/or paracrine signaling via locally generated SPMs in the vasculature may represent a novel homeostatic mechanism of relevance to vascular health and disease.-Chatterjee, A., Komshian, S., Sansbury, B. E., Wu, B., Mottola, G., Chen, M., Spite, M., Conte, M. S. Biosynthesis of proresolving lipid mediators by vascular cells and tissues.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Células Endoteliais/metabolismo , Metabolismo dos Lipídeos/fisiologia , Miócitos de Músculo Liso/metabolismo , Anticorpos , Araquidonato 5-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/metabolismo , Células Cultivadas , Citocinas/metabolismo , Ácidos Docosa-Hexaenoicos/genética , Ácidos Docosa-Hexaenoicos/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Inflamação/metabolismo , Leucócitos/fisiologia , Estrutura Molecular , Transporte Proteico/fisiologia , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lipoxinas/genética , Receptores de Lipoxinas/metabolismo
6.
Brain Res ; 1659: 113-120, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28089662

RESUMO

Paraplegia caused by spinal cord ischemia is a severe complication following surgeries in the thoracic aneurysm. HMGB1 has been recognized as a key mediator in spinal inflammatory response after spinal cord injury. Electroacupuncture (EA) pretreatment could provide neuroprotection against cerebral ischemic injury through inhibition of HMGB1 release. Therefore, the present study aims to test the hypothesis that EA pretreatment protects against spinal cord ischemia-reperfusion (I/R) injury via inhibition of HMGB1 release. Animals were pre-treated with EA stimulations 30min daily for 4 successive days, followed by 20-min spinal cord ischemia induced by using a balloon catheter placed into the aorta. We found that spinal I/R significantly increased mRNA and cytosolic protein levels of HMGB1 after reperfusion in the spinal cord. The EA-pretreated animals displayed better motor performance after reperfusion along with the decrease of apoptosis, HMGB1, TNF-α and IL-1ß expressions in the spinal cord, whereas these effects by EA pretreatment was reversed by rHMGB1 administration. Furthermore, EA pretreatment attenuated the down-regulation of LXA4 receptor (ALX) expression induced by I/R injury, while the decrease of HMGB1 release in EA-pretreated rats was reversed by the combined BOC-2 (an inhibitor of LXA4 receptor) treatment. In conclusion, EA pretreatment may promote spinal I/R injury through the inhibition of HMGB1 release in a LXA4 receptor-dependent manner. Our data may represent a new therapeutic technique for treating spinal cord ischemia-reperfusion injury.


Assuntos
Eletroacupuntura , Proteína HMGB1/metabolismo , Receptores de Lipoxinas/metabolismo , Traumatismo por Reperfusão/terapia , Isquemia do Cordão Espinal/terapia , Medula Espinal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Modelos Animais de Doenças , Interleucina-1beta/metabolismo , Masculino , Neurotransmissores/farmacologia , Oligopeptídeos/farmacologia , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores de Lipoxinas/antagonistas & inibidores , Recuperação de Função Fisiológica/fisiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Isquemia do Cordão Espinal/metabolismo , Isquemia do Cordão Espinal/patologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
7.
Can J Physiol Pharmacol ; 95(4): 340-348, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28060522

RESUMO

There is evidence for a relationship between inflammation and seizures because epilepsy can be caused by or result in inflammation. This study aimed to investigate the effect of aspirin and (or) omega-3 polyunsaturated fatty acids (PUFAs) on seizure activity and neurodegeneration in pentylenetetrazole (PTZ)-kindled rats focusing on their effect on corticohippocampal production of lipoxin A4 (LXA4) and expression of formyl peptide receptor-like 1 (FPRL1) receptors. Male rats were injected with PTZ (35 mg/kg, i.p.) 3 times per week for a total of 15 doses. Rats were treated daily with aspirin (20 mg/kg, i.p.), omega-3 PUFAs (85 mg/kg, p.o.), or a combination of them for 35 days. Both LXA4 level and expression of FPRL1 receptor in the cortices and hippocampi of rats' brains were greater in PTZ-kindled rats compared to a saline control group. Cotreatment with aspirin and (or) omega-3 PUFAs reduced convulsive behaviour; reduced levels of LXA4, interleukin-1ß, and nuclear factor-κB; and showed a lower percentage of corticohippocampal degenerative cells compared to PTZ-kindled rats. The combination of the 2 therapeutic agents did not provide significant improvement in comparison with the monotherapies. These findings suggest the use of aspirin or omega-3 PUFAs may delay the development of seizures and provide neuroprotection in a clinical setting.


Assuntos
Aspirina/uso terapêutico , Epilepsia/prevenção & controle , Ácidos Graxos Ômega-3/uso terapêutico , Lipoxinas/metabolismo , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Receptores de Lipoxinas/metabolismo , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Regulação para Baixo , Quimioterapia Combinada , Epilepsia/induzido quimicamente , Epilepsia/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Interleucina-1beta/metabolismo , Masculino , NF-kappa B/metabolismo , Pentilenotetrazol/toxicidade , Ratos , Receptores de Formil Peptídeo/metabolismo
8.
Sci Transl Med ; 8(353): 353ra111, 2016 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-27559094

RESUMO

Resolution of inflammation is a finely regulated process mediated by specialized proresolving lipid mediators (SPMs), including docosahexaenoic acid (DHA)-derived resolvins and maresins. The immunomodulatory role of SPMs in adaptive immune cells is of interest. We report that D-series resolvins (resolvin D1 and resolvin D2) and maresin 1 modulate adaptive immune responses in human peripheral blood lymphocytes. These lipid mediators reduce cytokine production by activated CD8(+) T cells and CD4(+) T helper 1 (TH1) and TH17 cells but do not modulate T cell inhibitory receptors or abrogate their capacity to proliferate. Moreover, these SPMs prevented naïve CD4(+) T cell differentiation into TH1 and TH17 by down-regulating their signature transcription factors, T-bet and Rorc, in a mechanism mediated by the GPR32 and ALX/FPR2 receptors; they concomitantly enhanced de novo generation and function of Foxp3(+) regulatory T (Treg) cells via the GPR32 receptor. These results were also supported in vivo in a mouse deficient for DHA synthesis (Elovl2(-/-)) that showed an increase in TH1/TH17 cells and a decrease in Treg cells compared to wild-type mice. Additionally, either DHA supplementation in Elovl2(-/-) mice or in vivo administration of resolvin D1 significantly reduced cytokine production upon specific stimulation of T cells. These findings demonstrate actions of specific SPMs on adaptive immunity and provide a new avenue for SPM-based approaches to modulate chronic inflammation.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Acetiltransferases/deficiência , Acetiltransferases/genética , Imunidade Adaptativa , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Diferenciação Celular , Elongases de Ácidos Graxos , Humanos , Inflamação/terapia , Mediadores da Inflamação/metabolismo , Interleucina-2/biossíntese , Metabolismo dos Lipídeos , Ativação Linfocitária , Camundongos , Camundongos Knockout , Receptores de Formil Peptídeo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lipoxinas/metabolismo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/citologia
9.
Am J Physiol Lung Cell Mol Physiol ; 310(11): L1053-61, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27084849

RESUMO

The specialized proresolution lipid mediator lipoxin A4 (LXA4) is abnormally produced in cystic fibrosis (CF) airways. LXA4 increases the CF airway surface liquid height and stimulates airway epithelial repair and tight junction formation. We report here a protective effect of LXA4 (1 nM) against tight junction disruption caused by Pseudomonas aeruginosa bacterial challenge together with a delaying action against bacterial invasion in CF airway epithelial cells from patients with CF and immortalized cell lines. Bacterial invasion and tight junction integrity were measured by gentamicin exclusion assays and confocal fluorescence microscopy in non-CF (NuLi-1) and CF (CuFi-1) bronchial epithelial cell lines and in primary CF cultures, grown under an air/liquid interface, exposed to either a clinical or laboratory strains of P. aeruginosa LXA4 delayed P. aeruginosa invasion and transepithelial migration in CF and normal bronchial epithelial cell cultures. These protective effects of LXA4 were inhibited by the ALX/FPR2 lipoxin receptor antagonist BOC-2. LXA4 prevented the reduction in mRNA biosynthesis and protein abundance of the tight junction protein ZO-1 and reduced tight junction disruption induced by P. aeruginsosa inoculation. In conclusion, LXA4 plays a protective role in bronchial epithelium by stimulating tight junction repair and by delaying and reducing the invasion of CF bronchial epithelial cells by P. aeruginsosa.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Fibrose Cística/tratamento farmacológico , Lipoxinas/farmacologia , Infecções por Pseudomonas/microbiologia , Junções Íntimas/metabolismo , Linhagem Celular , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Avaliação Pré-Clínica de Medicamentos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Expressão Gênica , Humanos , Infecções por Pseudomonas/prevenção & controle , Pseudomonas aeruginosa/fisiologia , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Mucosa Respiratória/microbiologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/microbiologia , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
10.
Reproduction ; 142(1): 15-28, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21490125

RESUMO

Inflammatory processes are central to reproductive events including ovulation, menstruation, implantation and labour, while inflammatory dysregulation is a feature of numerous reproductive pathologies. In recent years, there has been much research into the endogenous mechanisms by which inflammatory reactions are terminated and tissue homoeostasis is restored, a process termed resolution. The identification and characterisation of naturally occurring pro-resolution mediators including lipoxins and annexin A1 has prompted a shift in the field of anti-inflammation whereby resolution is now observed as an active process, triggered as part of a normal inflammatory response. This review will address the process of resolution, discuss available evidence for expression of pro-resolution factors in the reproductive tract and explore possible roles for resolution in physiological reproductive processes and associated pathologies.


Assuntos
Doenças dos Genitais Femininos/imunologia , Genitália Feminina/imunologia , Inflamação/metabolismo , Reprodução , Animais , Anexina A1/metabolismo , Anti-Inflamatórios/uso terapêutico , Eicosanoides/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Feminino , Doenças dos Genitais Femininos/tratamento farmacológico , Doenças dos Genitais Femininos/metabolismo , Genitália Feminina/efeitos dos fármacos , Genitália Feminina/metabolismo , Glucocorticoides/metabolismo , Homeostase , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Terapia de Alvo Molecular , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
J Immunol ; 186(4): 2087-94, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21228351

RESUMO

The hepatitis C virus (HCV) nonstructural 5A, a phosphorylated zinc metalloprotein, is an essential component of the HCV replication complex. An amphipathic α-helical peptide (HCV peptide [C5A]) derived from nonstructural 5A membrane anchor domain possesses potent anti-HCV and anti-HIV activity in vitro. In this study, we aimed to investigate the potential of HCV peptide (C5A) to regulate host immune responses. The capacity of HCV peptide (C5A) in vitro to induce migration and calcium mobilization of human phagocytes and chemoattractant receptor-transfected cells was investigated. The recruitment of phagocytes in vivo induced by HCV peptide (C5A) and its adjuvant activity were examined. The results revealed that HCV peptide (C5A) was a chemoattractant and activator of human phagocytic leukocytes by using a G-protein coupled receptor, namely formyl peptide receptor. In mice, HCV peptide (C5A) induced massive phagocyte infiltration after injection in the air pouch or the s.c. region. HCV peptide (C5A) also acted as an immune adjuvant by enhancing specific T cell responses to Ag challenge in mice. Our results suggest that HCV peptide (C5A) derived from HCV regulates innate and adaptive immunity in the host by activating the formyl peptide receptor.


Assuntos
Fragmentos de Peptídeos/fisiologia , Fagócitos/imunologia , Fagócitos/metabolismo , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Proteínas não Estruturais Virais/fisiologia , Imunidade Adaptativa , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Imunidade Inata , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/virologia , Fragmentos de Peptídeos/química , Fagócitos/virologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Receptores de Formil Peptídeo/fisiologia , Receptores de Lipoxinas/fisiologia , Proteínas não Estruturais Virais/química
12.
Eur J Immunol ; 37(7): 1966-77, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17559171

RESUMO

The objective was to evaluate which receptors house dust mite (HDM) and birch pollen extracts engage to activate human eosinophils. Chemotaxis and degranulation were studied in eosinophils pretreated with pertussis toxin and other antagonists of G protein-coupled receptors, e.g. the formyl peptide receptor (FPR), CC chemokine receptor 3 (CCR3) and leukotriene receptor B4 (LTB(4)R). Inhibition of the FPR as well as desensitization of the receptor rendered eosinophils anergic to activation by the allergens. Blockade of CCR3 or LTB(4)R did not affect eosinophilic reactivity. It was determined by PCR that human eosinophils express the FPR family members FPR and FPR-like 1 (FPRL1). HDM, unlike birch pollen, evoked calcium fluxes in HL-60 cells transfected with FPR or FPRL1. Although both allergens gave rise to calcium transients in neutrophils, which also express FPR and FPRL1, only the HDM response was decreased by the FPR antagonist. Moreover, neutrophils migrated toward HDM but not to birch pollen. Eosinophils pretreated with inhibitors of MAPK p38, ERK1/2 or protein kinase C exhibited diminished responsiveness to the aeroallergens. This study indicates that FPR and FPRL1 mediate the activation of eosinophils by HDM, whereas birch pollen employs other pathways shared with FPR to activate human eosinophils.


Assuntos
Antígenos de Dermatophagoides/imunologia , Eosinófilos/imunologia , Receptores de Formil Peptídeo/imunologia , Receptores de Lipoxinas/imunologia , Transdução de Sinais/imunologia , Animais , Betula/imunologia , Cálcio/metabolismo , Quimiotaxia de Leucócito/imunologia , Eosinófilos/metabolismo , Células HL-60 , Humanos , Pólen/imunologia , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , Receptores CCR3 , Receptores de Quimiocinas/imunologia , Receptores de Quimiocinas/metabolismo , Receptores de Formil Peptídeo/metabolismo , Receptores do Leucotrieno B4/imunologia , Receptores do Leucotrieno B4/metabolismo , Receptores de Lipoxinas/metabolismo , Transfecção
13.
J Immunol ; 174(8): 5033-9, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15814734

RESUMO

Acute lung injury (ALI) is a severe illness with excess mortality and no specific therapy. In its early exudative phase, neutrophil activation and accumulation in the lung lead to hypoxemia, widespread tissue damage, and respiratory failure. In clinical trials, inhibition of proinflammatory mediators has not proven effective. In this study, we pursued a new investigative strategy that emphasizes mediators promoting resolution from lung injury. A new spontaneously resolving experimental murine model of ALI from acid aspiration was developed to identify endogenous proresolving mechanisms. ALI increased cyclooxygenase 2 (COX-2) expression in murine lung. Selective pharmacologic inhibition or gene disruption of COX-2 blocked resolution of ALI. COX-2-derived products increased levels of the proresolving lipid mediators lipoxin A4 (LXA4) and, in the presence of aspirin, 15-epi-LXA4. Both LXA4 and 15-epi-LXA4 interact with the LXA4 receptor (ALX) to mediate anti-inflammatory actions. ALX expression was markedly induced by acid injury and transgenic mice with increased ALX expression displayed dramatic protection from ALI. Together, these findings indicate a protective role in ALI for COX-2-derived mediators, in part via enhanced lipoxin signaling, and carry potential therapeutic implications for this devastating clinical disorder.


Assuntos
Lesão Pulmonar , Pulmão/enzimologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , DNA Complementar/genética , Modelos Animais de Doenças , Expressão Gênica , Humanos , Ácido Clorídrico/toxicidade , Mediadores da Inflamação/metabolismo , Lipoxinas/biossíntese , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Transgênicos , Prostaglandina-Endoperóxido Sintases/genética , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/genética , Receptores de Lipoxinas/metabolismo , Transdução de Sinais
14.
Br J Pharmacol ; 141(1): 37-46, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14662730

RESUMO

1. Human formyl peptide-receptor-like-1 (FPRL-1) is a promiscuous G protein-coupled receptor (GPCR), and belongs to a chemoattractant receptor family protein. This receptor has been reported to interact with various host-derived peptides and lipids involved in inflammatory responses. We described here, a novel role for FPRL-1 as a high-affinity beta-chemokine receptor for an N-terminally truncated form of the CKbeta8 (CCL23/MPIF-1) splice variant CKbeta8-1 (22-137 aa). 2. RT-PCR analysis of mRNA derived from human tissues and cells revealed a predominant expression of FPRL-1 in inflammatory cells, particularly in neutrophils. 3. Intracellular calcium mobilisation assay, used as screening tool, in recombinant Chinese hamster ovary (CHO-K1) and human embryonic kidney (HEK293s) cells coexpressing FPRL-1 and Galpha(16), demonstrated FPRL-1 is a functional high-affinity receptor for CKbeta8-1 (46-137 aa, sCKbeta8-1), with pEC(50) values of 9.13 and 8.85, respectively. 4. The FPRL-1 activation in CHO-K1 cells is mediated by Galpha(i)/Galpha(o) proteins, as assessed by pertussis toxin sensitivity and inhibition of forskolin-induced cyclic AMP accumulation. 5. Binding experiments were performed with a radio-iodinated synthetic peptide, [(125-)I]-WKYMVm, a known potent FPRL-1 agonist. CHO-K1 cell membranes expressing FPRL-1 bound [(125-)I]-WKYMVm with a K(d) value of 9.34. Many known FPRL-1 agonists were tested and sCKbeta8-1 was the most effective nonsynthetic ligand in displacing the radiolabelled agonist, with a pIC(50) of 7.97. 6. The functional significance of sCKbeta8-1 interaction with FPRL-1 was further demonstrated by the activation of polymorphonuclear leukocytes (PMNs) calcium mobilisation and chemotaxis. These interactions were shown to be via FPRL-1 by specific blockade of PMNs activation in the presence of an FPRL-1 antibody.


Assuntos
Quimiocinas CC/química , Quimiocinas CC/farmacologia , Receptores de Formil Peptídeo/efeitos dos fármacos , Receptores de Lipoxinas/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Células CHO , Cálcio/metabolismo , Movimento Celular/efeitos dos fármacos , Quimiocinas CC/metabolismo , Quimiotaxia/efeitos dos fármacos , Cricetinae , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica , Humanos , Radioisótopos do Iodo/metabolismo , Rim/citologia , Rim/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/genética , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores Acoplados a Proteínas G , Receptores de Lipoxinas/genética , Receptores de Lipoxinas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA