Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
J Phys Chem B ; 127(29): 6479-6486, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37428488

ABSTRACT

Chronic inflammation contributes to several diseases, but its resolution is driven by specialized pro-resolving mediators (SPM) such as resolvin D1 (RvD1) and its epimer aspirin-triggered resolvin D1 (AT-RvD1), both biosynthesized from ω-3 fatty docosahexaenoic acid (DHA). RvD1 and AT-RvD1 have anti-inflammatory and pro-resolution potentials, and their effects could be mediated by formyl peptide receptor type 2 receptor ALX/FPR2, a G-protein-coupled receptor (GPCR). In this work, we performed 44 µs of molecular dynamics simulations with two complexes: FPR2@AT-RvD1 and FPR2@RvD1. Our results show the following: (i) in the AT-RvD1 simulations, the ALX/FPR2 receptor remained in the active state in 62% of the frames, while in the RVD1 simulations, the receptor remained in the active state in 74% of the frames; (ii) two residues, R201 and R205, of ALX/FPR2 appear, establishing interactions with both resolvins in all simulations (22 in total); (iii) RvD1 hydrogen bonds with R201 and R205 presented higher frequency than AT-RvD1; and (iv) residues R201 and R205 are the two receptor hotspots, demonstrated by the binding free calculations. Such results show that the ALX/FPR2 receptor remained in the active state for longer in the FPR2@RvD1 simulations than in the FPR2@AT-RvD1 simulations.


Subject(s)
Molecular Dynamics Simulation , Receptors, Formyl Peptide , Humans , Receptors, Formyl Peptide/metabolism , Stereoisomerism , Inflammation/metabolism , Aspirin , Receptors, Lipoxin/physiology
2.
J Pharmacol Sci ; 148(1): 56-64, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924130

ABSTRACT

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.


Subject(s)
Acute Kidney Injury/genetics , Acute Kidney Injury/prevention & control , Chalcones/pharmacology , Chalcones/therapeutic use , Cisplatin/adverse effects , Macrophages/metabolism , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Animals , Cells, Cultured , Chalcones/isolation & purification , Gene Expression/drug effects , Glycyrrhiza/chemistry , Inflammation , Male , Mice, Inbred C57BL , Molecular Targeted Therapy , Phytotherapy , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Receptors, Lipoxin/physiology , Up-Regulation/drug effects
3.
Mol Med Rep ; 24(6)2021 12.
Article in English | MEDLINE | ID: mdl-34608500

ABSTRACT

Recurrent spontaneous abortion (RSA) effects both the physical and mental health of women of reproductive age. Trophoblast dysfunction may result in RSA due to shallow placental implantation. The mechanisms underlying formyl peptide receptor 2 (FPR2) on the biological functions of trophoblasts remain to be elucidated. The present study aimed to explore the potential functions of FPR2, a G protein­coupled receptor, in placental trophoblasts. The location and expression levels of FPR2 in the villi tissue of patients with RSA were detected using immunohistochemical staining, reverse transcription­quantitative PCR and western blotting. Following the transfection of small interfering RNA targeting FPR2 in HTR­8/SVneo cells, a Cell Counting Kit­8 assay was used to determine the levels of cell viability. Flow cytometry was used to examine the levels of cell apoptosis and gap closure and Transwell assays were carried out to evaluate the levels of cell migration and invasion. A tube formation assay was performed to detect the levels of capillary­like structure formation. Western blotting was used to detect the expression levels of proteins in the associated signaling pathways. The expression of FPR2 was present in villi trophoblasts and was markedly increased in patients with RSA. The levels of trophoblast invasion, migration and tube formation were markedly increased following FPR2 knockdown, whereas the levels of apoptosis were markedly decreased. In addition, FPR2 knockdown caused an increase in the phosphorylation levels of AKT and PI3K. Thus, FPR2 may be involved in the regulation of trophoblast function via the PI3K/AKT signaling pathway. The results of the present study provided a theoretical basis for the use of FPR2 as a target for the treatment of trophoblast­associated diseases, such as RSA.


Subject(s)
Abortion, Habitual/physiopathology , Chorionic Villi/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Trophoblasts/metabolism , Apoptosis , Cell Line , Cell Movement , Cell Survival , Female , Gene Knockdown Techniques/methods , Humans , Pregnancy , RNA, Small Interfering/metabolism , Signal Transduction
4.
Cell Death Dis ; 11(8): 653, 2020 08 08.
Article in English | MEDLINE | ID: mdl-32811815

ABSTRACT

Radiation therapy is an important modality in the treatment of lung cancer, but it can lead to radiation pneumonitis, and eventually radiation fibrosis. To date, only few available drugs can effectively manage radiation-induced pulmonary fibrosis. Lipoxins are endogenous molecules exhibit anti-inflammatory and pro-resolving effects. These molecules play a vital role in reducing excessive tissue injury and chronic inflammation; however, their effects on radiation-induced lung injury (RILI) are unknown. In this study, we investigated the effects of lipoxin A4 (LXA4) on RILI using our specialized small-animal model of RILI following focal-ablative lung irradiation (IR). LXA4 significantly inhibited immune-cell recruitment and reduced IR-induced expression of pro-inflammatory cytokines and fibrotic proteins in the lung lesion sites. In addition, micro-CT revealed that LXA4 reduced IR-induced increases in lung consolidation volume. The flexiVentTM assays showed that LXA4 significantly reversed IR-induced lung function damage. Moreover, LXA4 downregulated the activities of NF-κB and the Smad-binding element promoters. The expression of FPR2, an LXA4 receptor, increased during the development of IR-induced pulmonary fibrosis, whereas silencing of endogenous LXA4 using an antagonist (WRW4) or FPR2 siRNA resulted in impaired development of pulmonary fibrosis in response to IR. Collectively, these data suggest that LXA4 could serve as a potent therapeutic agent for alleviating RILI.


Subject(s)
Lipoxins/metabolism , Pulmonary Fibrosis/metabolism , Receptors, Formyl Peptide/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Fibrosis/metabolism , Humans , Lipoxins/physiology , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/physiopathology , Radiation , Radiotherapy/adverse effects , Receptor Cross-Talk/physiology , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/metabolism , Receptors, Lipoxin/physiology , Signal Transduction/physiology , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
5.
Lab Invest ; 98(6): 715-733, 2018 06.
Article in English | MEDLINE | ID: mdl-29467458

ABSTRACT

Inflammation always accompanies infection during sepsis. Mitochondrial dysfunction and the role of reactive oxygen species (ROS) produced by mitochondria have been proposed in the pathogenesis of sepsis. Maresins have protective and resolving effects in experimental models of infection. In the present study, we investigated the effects of maresin 1 (MaR1) on mitochondrial function in cecal ligation and puncture (CLP)-induced sepsis and sepsis patients to identify mechanisms underlying maresin 1-mediated stimulation of ROS in mitochondria. We found that treatment with MaR1 significantly inhibited production of cytokines, decreased bacterial load in the peritoneal lavage fluid, reduced the number of neutrophils, decreased lactic acid level and upregulated cyclic AMP (cAMP) concentration, with the outcome of decreased lung injury in CLP-induced sepsis in mice. The effects of MaR1 on downregulation nitric oxide (NOX) activity, improvement CAT and SOD activity to inhibit ROS production in mitochondria was dependent on lipoxin receptor (ALX) and cAMP. Survival rates were significantly increased after the treatment of mice with MaR1. In BMDM stimulated with LPS, MaR1 inhibited the ROS production, downregulated enzyme activity, reduced mtO2 production, increased mitochondrial membrane potential, improved adenosine triphosphate (ATP) content and mitochondrial DNA (mtDNA) copy number. Finally, the effects of MaR1 on ROS production in the blood of healthy volunteers stimulated with LPS or sepsis patients were associated with ALX and cAMP. Taken together, these data suggest that treatment with MaR1 could attenuate mitochondrial dysfunction during sepsis through regulating ROS production.


Subject(s)
Cyclic AMP/physiology , Docosahexaenoic Acids/pharmacology , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Receptors, Lipoxin/physiology , Sepsis/drug therapy , Signal Transduction/physiology , Animals , Catalase/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/physiology , Nitric Oxide/physiology , Oligopeptides/pharmacology , Sepsis/immunology , Sepsis/mortality , Signal Transduction/drug effects
6.
Biochim Biophys Acta ; 1853(1): 192-200, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25447672

ABSTRACT

Pathogenic Staphylococcus aureus strains produce N-formylmethionyl containing peptides, of which the tetrapeptide fMIFL is a potent activator of the neutrophil formyl peptide receptor 1 (FPR1) and the PSMα2 peptide is a potent activator of the closely related FPR2. Variants derived from these two peptide activators were used to disclose the structural determinants for receptor interaction. Removal of five amino acids from the C-terminus of PSMα2 gave rise to a peptide that had lost the receptor-independent neutrophil permeabilizing effect, whereas neutrophil activation capacity as well as its preference for FPR2 was retained. Shorter peptides, PSMα21-10 and PSMα21-5, activate neutrophils, but the receptor preference for these peptides was switched to FPR1. The fMIFL-PSM5-16 peptide, in which the N-terminus of PSMα21-16 was replaced by the sequence fMIFL, was a dual agonist for FPR1/FPR2, whereas fMIFL-PSM5-10 preferred FPR1 to FPR2. Further, an Ile residue was identified as a key determinant for interaction with FPR2. A chimeric receptor in which the cytoplasmic tail of FPR1 was replaced by the corresponding part of FPR2 lost the ability to recognize FPR1 agonists, but gained function in relation to FPR2 agonists. Taken together, our data demonstrate that the C-terminus of the PSMα2 peptide plays a critical role for its cytotoxicity, but is not essential for the receptor-mediated pro-inflammatory activity. More importantly, we show that the amino acids present in the C-terminus, which are not supposed to occupy the agonist-binding pocket in the FPRs, are of importance for the choice of receptor.


Subject(s)
N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation/drug effects , Receptors, Formyl Peptide/chemistry , Receptors, Lipoxin/chemistry , Humans , Ligands , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/agonists , Receptors, Lipoxin/physiology , Structure-Activity Relationship
7.
Nat Rev Rheumatol ; 9(10): 595-603, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23958797

ABSTRACT

Glucocorticoids have broad-ranging and powerful anti-inflammatory and immunomodulatory effects. Unsurprisingly, therefore, glucocorticoids are widely and persistently used to treat a large number of inflammatory diseases, including rheumatoid arthritis (RA), despite the well-described adverse effects of these drugs. Annexin A1 is a glucocorticoid-induced molecule that is known to replicate many of the described anti-inflammatory effects of glucocorticoids. In addition to the well-documented roles of this protein in neutrophil function, emerging evidence suggests that annexin A1 is involved in the modulation of T-cell function and the adaptive immune responses relevant to RA. Interest in annexin A1 was renewed after the delineation of the receptors for this protein. This breakthrough also led to advances in our understanding of anti-inflammatory annexin A1 mimetic peptides and agonistic compounds targeting these receptors, particularly those specific for the receptor N-formyl peptide receptor 2 (FPR2). Herein, we review the current knowledge of the biological activities of annexin A1 and their relevance to RA pathogenesis. We also discuss the potential of annexin A1 mimics and strategies aimed at potentiating annexin A1 signalling to become viable approaches to minimizing glucocorticoid use in RA and other inflammatory disorders.


Subject(s)
Annexin A1/physiology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/physiopathology , Glucocorticoids/therapeutic use , Adaptive Immunity/physiology , Glucocorticoids/pharmacology , Humans , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , T-Lymphocytes/pathology
8.
J Biol Chem ; 288(12): 8258-8268, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23386607

ABSTRACT

LL-37 is an antimicrobial peptide produced by human cells that can down-regulate the lipopolysaccharide-induced innate immune responses and up-regulate double-stranded (ds) RNA-induced innate responses through Toll-like receptor 3 (TLR3). The murine LL-37 ortholog, mCRAMP, also inhibited lipopolysaccharide-induced responses, but unlike LL-37, it inhibited viral-induced responses in mouse cells. A fluorescence polarization assay showed that LL-37 was able to bind dsRNA better than mCRAMP. In the human lung epithelial cell line BEAS-2B, LL-37, but not mCRAMP, colocalized with TLR3, and the colocalization was increased in the presence of dsRNA. The presence of poly(I:C) increased the accumulation of LL-37 in Rab5 endosomes. Signaling by cells induced with both LL-37 and poly(I:C) was sensitive to inhibitors that affect clathrin-independent trafficking, whereas signaling by poly(I:C) alone was not, suggesting that the LL-37-poly(I:C) complex trafficked to signaling endosomes by a different mechanism than poly(I:C) alone. siRNA knockdown of known LL-37 receptors identified that FPRL1 was responsible for TLR3 signaling induced by LL-37-poly(I:C). These results show that LL-37 and mCRAMP have different activities in TLR3 signaling and that LL-37 can redirect trafficking of poly(I:C) to effect signaling by TLR3 in early endosomes in a mechanism that involves FPRL1.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Cathelicidins/physiology , Poly I-C/pharmacology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Amino Acid Sequence , Animals , Cell Line , Endocytosis/drug effects , Endosomes/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mice , Molecular Sequence Data , Protein Binding , Protein Transport , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Signal Transduction , Toll-Like Receptor 3/metabolism , beta-Cyclodextrins/pharmacology
9.
Atherosclerosis ; 227(1): 72-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23340376

ABSTRACT

Serum amyloid A (SAA) is an acute phase protein whose expression increases markedly during bacterial infection, tissue damage, and inflammation. The potential beneficial roles of SAA include its involvement in the reverse cholesterol transport and possibly extracellular lipid deposition at sites of inflammation and tissue repair. It is an attractive therapeutic target for the treatment of atherosclerosis. Peroxisome proliferator-activated receptor γ (PPARγ) plays a major regulatory role in adipogenesis, and the expression of genes involved in lipid metabolism. Activation of PPARγ leads to multiple changes in gene expression, some of which are believed to be atherogenic while others are antiatherogenic. In this study, we demonstrated that SAA upregulated COX-2 expression and induced PPARγ activity through NF-кB pathway. The effect of SAA on NF-кB activity is mediated by FPRL-1 and TLR4.


Subject(s)
NF-kappa B/physiology , PPAR gamma/metabolism , Serum Amyloid A Protein/physiology , Cyclooxygenase 2 , Flavonoids/pharmacology , Hep G2 Cells , Humans , Lipoproteins, HDL/physiology , NF-kappa B/antagonists & inhibitors , NF-kappa B/drug effects , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Toll-Like Receptor 4/physiology
10.
J Immunol ; 189(2): 629-37, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706076

ABSTRACT

The neutrophil formyl peptide receptors, FPR1 and FPR2, play critical roles for inflammatory reactions, and receptor-specific antagonists/inhibitors can possibly be used to facilitate the resolution of pathological inflammatory reactions. A 10-aa-long rhodamine-linked and membrane-permeable peptide inhibitor (PBP10) has such a potential. This FPR2 selective inhibitor adopts a phosphatidylinositol 4,5-bisphosphate-binding sequence in the cytoskeletal protein gelsolin. A core peptide, RhB-QRLFQV, is identified that displays inhibitory effects as potent as the full-length molecule. The phosphatidylinositol 4,5-bisphosphate-binding capacity of PBP10 was not in its own sufficient for inhibition. A receptor in which the presumed cytoplasmic signaling C-terminal tail of FPR2 was replaced with that of FPR1 retained the PBP10 sensitivity, suggesting that the tail of FPR2 was not on its own critical for inhibition. This gains support from the fact that the effect of cell-penetrating lipopeptide (a pepducin), suggested to act primarily through the third intracellular loop of FPR2, was significantly inhibited by PBP10. The third intracellular loops of FPR1 and FPR2 differ in only two amino acids, but an FPR2 mutant in which these two amino acids were replaced by those present in FPR1 retained the PBP10 sensitivity. In summary, we conclude that the inhibitory activity on neutrophil function of PBP10 is preserved in the core sequence RhB-QRLFQV and that neither the third intracellular loop of FPR2 nor the cytoplasmic tail of the receptor alone is responsible for the specific inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Gelsolin/chemistry , Gelsolin/physiology , Peptides/chemistry , Peptides/physiology , Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/chemistry , Receptors, Lipoxin/physiology , Amino Acid Sequence , Cell Membrane Permeability/immunology , Dose-Response Relationship, Immunologic , Gelsolin/metabolism , HL-60 Cells , Humans , Molecular Sequence Data , Neutrophil Activation/immunology , Peptides/metabolism , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism
11.
Med Hypotheses ; 79(1): 101-3, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22541858

ABSTRACT

Atherosclerosis is a chronic inflammatory disease and the number one cause of mortality worldwide. The fundamental causes of atherosclerosis have not been precisely delineated, although pathogenesis clearly involves endothelial dysfunction and both innate and adaptive immunity. Recent evidence suggests that formyl peptide receptor 2 (FPR2), a G protein-coupled receptor (GPCR), mediates a range of inflammatory responses including superoxide production in neutrophils, chemotaxis of monocytes and neutrophils, CCL2 production in endothelial cells (ECs) and monocytes, and increased CXCL8 expression in neutrophils, which are all related with atherogenesis. Therefore, we propose that FPR2 may play a pathogenic role in atherogenesis.


Subject(s)
Atherosclerosis/physiopathology , Leukocytes/physiology , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Humans , Monocytes/metabolism , Neutrophils/metabolism , Superoxides/metabolism
12.
Arterioscler Thromb Vasc Biol ; 32(8): 1970-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22499990

ABSTRACT

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.


Subject(s)
Docosahexaenoic Acids/physiology , Inflammation/pathology , Neutrophils/physiology , Animals , Cell Movement , Humans , Male , Mice , Receptors, Formyl Peptide/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Lipoxin/physiology
13.
Am J Pathol ; 180(5): 2018-27, 2012 May.
Article in English | MEDLINE | ID: mdl-22449948

ABSTRACT

Resolution of acute inflammation is an active process that involves the biosynthesis of specialized proresolving lipid mediators. Among them, resolvin D1 (RvD1) actions are mediated by two G protein-coupled receptors (GPCRs), ALX/FPR2 and GPR32, that also regulate specific microRNAs (miRNAs) and their target genes in novel resolution circuits. We report the ligand selectivity of RvD1 activation of ALX/FPR2 and GPR32. In addition to RvD1, its aspirin-triggered epimer and RvD1 analogs each dose dependently and effectively activated ALX/FPR2 and GPR32 in GPCR-overexpressing ß-arrestin systems using luminescence and electric cell-substrate impedance sensing. To corroborate these findings in vivo, neutrophil infiltration in self-limited peritonitis was reduced in human ALX/FPR2-overexpressing transgenic mice that was further limited to 50% by RvD1 treatment with as little as 10 ng of RvD1 per mouse. Analysis of miRNA expression revealed that RvD1 administration significantly up-regulated miR-208a and miR-219 in exudates isolated from ALX/FPR2 transgenic mice compared with littermates. Overexpression of miR-208a in human macrophages up-regulated IL-10. In comparison, in ALX/FPR2 knockout mice, RvD1 neither significantly reduced leukocyte infiltration in zymosan-induced peritonitis nor regulated miR-208a and IL-10 in these mice. Together, these results demonstrate the selectivity of RvD1 interactions with receptors ALX/FPR2 and GPR32. Moreover, they establish a new molecular circuit that is operative in the resolution of acute inflammation activated by the proresolving mediator RvD1 involving specific GPCRs and miRNAs.


Subject(s)
Docosahexaenoic Acids/physiology , Inflammation/metabolism , MicroRNAs/genetics , Receptors, G-Protein-Coupled/physiology , Acute Disease , Animals , Arrestins/metabolism , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Dose-Response Relationship, Drug , Electric Impedance , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Inflammation/genetics , Interleukin-10/biosynthesis , Interleukin-10/genetics , Ligands , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/physiology , Peritonitis/drug therapy , Peritonitis/metabolism , Peritonitis/pathology , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Up-Regulation/drug effects , beta-Arrestins
14.
Br J Anaesth ; 107(6): 948-58, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21990306

ABSTRACT

BACKGROUND: Annexin 1 (ANXA1) has analgesic effects in inflammatory pain. We aimed to investigate the anti-nociceptive role of ANXA1, at the dorsal root ganglion (DRG) level, through an interaction with formyl-peptide-receptor-like 1 (FPR2/ALX). METHODS: Inflammatory pain was evoked by injecting complete Freund's adjuvant (CFA, 50 µl) into the hindpaw of male Sprague-Dawley rats. The distribution of ANXA1 and FPR2/ALX in L4/5 DRGs was evaluated by immunofluorescence. The expression of ANXA1 was measured by western blot. The involvement of FPR2/ALX in the anti-nociception of ANXA1 was investigated by thermal (irradiant heat) and mechanical (von Frey filament) pain tests with intrathecal (i.t.) ANXA1-derived peptide (Anxa1(2-26)), FPR2/ALX agonist 5(S)-6(R)-7-trihydroxyheptanoic-acid-methyl-ester (BML-111), and antagonist N-t-Boc-Phe-Leu-Phe-Leu-Phe (Boc1). RESULTS: ANXA1 and FPR2/ALX localized in the satellite glial cells and neurones in L4/5 DRGs. CFA treatment (n=20) increased ANXA1 expression in L4/5 DRGs within 7 days (P<0.01). I.T. Anxa1(2-26) (20 and 100 µg µl(-1)) and BML-111 (10 and 100 nmol) reduced CFA-induced thermal and mechanical nociception within 48 h (n=40) (P<0.05). However, i.t. Boc1 10 µg intensified inflammatory pain (P<0.05) and reversed the anti-nociceptive effect of Anxa1(2-26) (n=25) (P<0.05). Moreover, ANXA1 expression increased in L4/5 DRGs after i.t. Anxa1(2-26) (20 µg µl(-1)) (P<0.05) and BML-111 (10 nmol) (P<0.01) but decreased after i.t. Boc1 (10 and 100 µg) alone (P<0.01) or Boc1 (10 µg) co-injection with Anxa1(2-26) (20 µg µl(-1)) (P<0.05). CONCLUSIONS: Endogenous ANXA1 expression at the DRG level is involved in CFA-induced inflammatory pain, and i.t. ANXA1 20 µg µl(-1) produces its anti-nociceptive effect through FPR2/ALX.


Subject(s)
Annexin A1/physiology , Ganglia, Spinal/physiology , Nociception , Pain/physiopathology , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Amino Acid Sequence , Animals , Annexin A1/analysis , Freund's Adjuvant/pharmacology , Inflammation/physiopathology , Male , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Receptors, Formyl Peptide/analysis , Receptors, Lipoxin/analysis
15.
J Immunol ; 186(4): 2087-94, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21228351

ABSTRACT

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.


Subject(s)
Peptide Fragments/physiology , Phagocytes/immunology , Phagocytes/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Viral Nonstructural Proteins/physiology , Adaptive Immunity , Amino Acid Sequence , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunity, Innate , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Peptide Fragments/chemistry , Phagocytes/virology , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Viral Nonstructural Proteins/chemistry
16.
Mol Pharmacol ; 79(1): 77-90, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20943772

ABSTRACT

N-Formyl peptide receptors (FPRs) are G protein-coupled receptors (GPCRs) involved in host defense and sensing cellular dysfunction. Thus, FPRs represent important therapeutic targets. In the present studies, we screened 32 ligands (agonists and antagonists) of unrelated GPCRs for their ability to induce intracellular Ca²+ mobilization in human neutrophils and HL-60 cells transfected with human FPR1, FPR2, or FPR3. Screening of these compounds demonstrated that antagonists of gastrin-releasing peptide/neuromedin B receptors (BB1/BB2) PD168368 [(S)-a-methyl-a-[[[(4-nitrophenyl)amino]carbonyl]amino]-N-[[1-(2-pyridinyl) cyclohexyl]methyl]-1H-indole-3-propanamide] and PD176252 [(S)-N-[[1-(5-methoxy-2-pyridinyl)cyclohexyl]methyl]-a-methyl-a-[[-(4-nitrophenyl)amino]carbonyl]amino-1H-indole-3-propanamide] were potent mixed FPR1/FPR2 agonists, with nanomolar EC50 values. Cholecystokinin-1 receptor agonist A-71623 [Boc-Trp-Lys(ε-N-2-methylphenylaminocarbonyl)-Asp-(N-methyl)-Phe-NH2] was also a mixed FPR1/FPR2 agonist, but with a micromolar EC50. Screening of 56 Trp- and Phe-based PD176252/PD168368 analogs and 41 related nonpeptide/nonpeptoid analogs revealed 22 additional FPR agonists. Most were potent mixed FPR1/FPR2/FPR3 agonists with nanomolar EC50 values for FPR2, making them among the most potent nonpeptide FPR2 agonists reported to date. In addition, these agonists were also potent chemoattractants for murine and human neutrophils and activated reactive oxygen species production in human neutrophils. Molecular modeling of the selected agonists using field point methods allowed us to modify our previously reported pharmacophore model for the FPR2 ligand binding site. This model suggests the existence of three hydrophobic/aromatic subpockets and several binding poses of FPR2 agonists in the transmembrane region of this receptor. These studies demonstrate that FPR agonists could include ligands of unrelated GPCR and that analysis of such compounds can enhance our understanding of pharmacological effects of these ligands.


Subject(s)
Indoles/pharmacology , Pyridines/pharmacology , Receptors, Bombesin/antagonists & inhibitors , Receptors, Formyl Peptide/agonists , Receptors, Lipoxin/agonists , Animals , Gastrin-Releasing Peptide/antagonists & inhibitors , Gastrin-Releasing Peptide/physiology , HL-60 Cells , Humans , Indoles/chemistry , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , Neutrophils/physiology , Pyridines/chemistry , Receptors, Bombesin/physiology , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology
17.
FASEB J ; 25(4): 1254-63, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21183593

ABSTRACT

The mechanisms used by the immune system to discriminate between pathogenic and commensal bacteria have remained largely unclear. Recently, we have shown that virulence of Staphylococcus aureus depends on secretion of phenol-soluble modulin (PSM) peptides that disrupt neutrophils at micromolar concentrations. Moreover, all S. aureus PSMs stimulate and attract neutrophils at nanomolar concentrations via interaction with the formyl-peptide receptor 2 (FPR2). Here, we demonstrate that FPR2 allows neutrophils to adjust their responses in relation to the aggressiveness of staphylococcal species, which differ largely in their capacity to infect or colonize humans and animals. PSM-related peptides were detected in all human and animal pathogenic staphylococci, but were absent from most commensal species. Three PSMß-like peptides produced by the serious human pathogen Staphylococcus lugdunensis were identified as the previously described S. lugdunensis-synergistic hemolysins (SLUSHs). SLUSHs attracted and stimulated human leukocytes in a FPR2-dependent manner, indicating that FPR2 is a general receptor for all PSM-like peptide toxins. Remarkably, the release of PSMs correlated closely with the apparent capacity of staphylococcal species to cause invasive infections and with their ability to activate FPR2. These findings suggest that the innate immune system may be able to respond in different ways to pathogenic or innocuous staphylococci by monitoring the presence of PSMs via FPR2.


Subject(s)
Bacterial Toxins/metabolism , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Staphylococcus/pathogenicity , Amino Acid Sequence , Animals , Bacterial Proteins/pharmacology , Chemotaxis, Leukocyte , HL-60 Cells , Hemolysin Proteins/pharmacology , Humans , Neutrophils/drug effects , Neutrophils/immunology , Staphylococcus aureus/pathogenicity , Staphylococcus lugdunensis/metabolism , Staphylococcus lugdunensis/pathogenicity , Virulence
18.
Pharmacology ; 84(5): 294-9, 2009.
Article in English | MEDLINE | ID: mdl-19816089

ABSTRACT

Rat gastric mucosal damage was induced by ischemia-reperfusion. The 5-lipoxygenase inhibitors MK886 and A63162, the 12-lipoxygenase inhibitor baicalein, the 15-lipoxygenase inhibitor PD146176 and the lipoxin (LX) A(4)/annexin 1 antagonist Boc1 increased mucosal damage in a dose-dependent manner. Low doses of these compounds, which have no effects on mucosal integrity, cause severe damage when combined with low doses of indomethacin, celecoxib or dexamethasone. 16,16-Dimethylprostaglandin (PG) E(2) and LXA(4) can replace each other in preventing mucosal injury induced by either cyclooxygenase or lipoxygenase inhibitors. The results suggest that not only cyclooxygenases, but also lipoxygenases have a role in limiting gastric mucosal damage during ischemia-reperfusion.


Subject(s)
Annexins/metabolism , Gastric Mucosa/drug effects , Lipoxygenase/physiology , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Reperfusion Injury/drug therapy , 16,16-Dimethylprostaglandin E2/pharmacology , Acetamides/pharmacology , Animals , Anti-Ulcer Agents/pharmacology , Celecoxib , Cyclooxygenase Inhibitors/pharmacology , Dexamethasone/pharmacology , Drug Synergism , Flavanones/pharmacology , Fluorenes/pharmacology , Gastric Mucosa/blood supply , Gastric Mucosa/pathology , Glucocorticoids/pharmacology , Indoles/pharmacology , Indomethacin/pharmacology , Lipoxygenase Inhibitors/pharmacology , Male , Oligopeptides/pharmacology , Phenyl Ethers , Prostaglandin Antagonists/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptors, Lipoxin/antagonists & inhibitors , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sulfonamides/pharmacology
19.
J Immunol ; 183(9): 5511-7, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19843937

ABSTRACT

We determined the expression of the formyl peptide receptor (FPR) family and the functional roles of the FPR family in NK cells. All tested human NK cells express two members of the FPR family (FPR1 and FPR2). The expression of FPR3 was noted to occur in a donor-specific manner. The stimulation of NK cells with FPR family-selective agonists (fMLF (N-formyl-Met-Leu-Phe), MMK-1, F2L, and WKYMVm (Trp-Lys-Tyr-Met-Val-d-Met)) elicited cytolytic activity in resting NK cells, but not in IL-2-activated NK cells; the cytolytic activity was not inhibited by pertussis toxin. The FPR family agonists also stimulated chemotactic migration of IL-2-activated NK cells, but not resting NK cells; the chemotactic migration was completely inhibited by pertussis toxin. WKYMVm stimulates ERK, p38 MAPK, and JNK activities in both resting and IL-2-activated NK cells. WKYMVm-induced chemotactic migration was partially inhibited by PD98059 (2'-amino-3'-methoxyflavone); however, the inhibition of JNK by its selective inhibitor (SP600125, anthra[1,9-cd]pyrazol-6(2H)-one) dramatically inhibited the WKYMVm-induced cytolytic activity. Furthermore, WKYMVm-induced chemotactic migration and cytolytic activity were partly inhibited by FPR family-selective antagonists (cyclosporin H and WRWWWW). Taken together, our findings indicate that human NK cells express functional members of the FPR family, and in turn the activation of the three members of the FPR receptor family elicit cytolytic activity in NK cells, thus suggesting that the receptors are potentially important therapeutic targets for the modulation of NK cell-mediated immune responses.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Multigene Family/immunology , Receptors, Formyl Peptide/biosynthesis , Chemotaxis, Leukocyte/immunology , Cytotoxicity, Immunologic , Humans , Lymphocyte Activation/immunology , Oligopeptides/agonists , Oligopeptides/biosynthesis , Oligopeptides/physiology , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/agonists , Receptors, Lipoxin/biosynthesis , Receptors, Lipoxin/physiology , Signal Transduction/immunology
20.
Nat Rev Immunol ; 9(1): 62-70, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19104500

ABSTRACT

Glucocorticoids are widely used for the management of inflammatory diseases. Their clinical application stems from our understanding of the inhibitory effect of the corticosteroid hormone cortisol on several components of the immune system. Endogenous and exogenous glucocorticoids mediate their multiple anti-inflammatory effects through many effector molecules. In this Opinion article, we focus on the role of one such effector molecule, annexin A1, and summarize the recent studies that provide insight into its molecular and pharmacological functions in immune responses. In addition, we propose a model in which glucocorticoids regulate the expression and function of annexin A1 in opposing ways in innate and adaptive immune cells to mediate the resolution of inflammation.


Subject(s)
Annexin A1/physiology , Glucocorticoids/physiology , Immune System/physiology , Inflammation/physiopathology , Animals , Annexin A1/deficiency , Annexin A1/genetics , Anti-Inflammatory Agents/therapeutic use , Circadian Rhythm , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Humans , Hydrocortisone/physiology , Immune System/drug effects , Immunity, Innate/drug effects , Lipoxins/physiology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Models, Immunological , Monocytes/immunology , Monocytes/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Formyl Peptide/deficiency , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...