Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Proc Natl Acad Sci U S A ; 117(14): 7971-7980, 2020 04 07.
Article En | MEDLINE | ID: mdl-32205444

Timely resolution of bacterial infections critically depends on phagocytosis of invading pathogens by polymorphonuclear neutrophil granulocytes (PMNs), followed by PMN apoptosis and efferocytosis. Here we report that bacterial DNA (CpG DNA) and mitochondrial DNA impair phagocytosis and attenuate phagocytosis-induced apoptosis in human PMNs through Toll-like receptor 9 (TLR9)-mediated release of neutrophil elastase and proteinase 3 and subsequent down-regulation of the complement receptor C5aR. Consistently, CpG DNA delays pulmonary clearance of Escherichia coli in mice and suppresses PMN apoptosis, efferocytosis, and generation of proresolving lipid mediators, thereby prolonging lung inflammation evoked by E. coli Genetic deletion of TLR9 renders mice unresponsive to CpG DNA. We also show that aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 (17-epi-RvD1) through the receptor ALX/FPR2 antagonize cues from CpG DNA, preserve C5aR expression, restore impaired phagocytosis, and redirect human PMNs to apoptosis. Treatment of mice with 15-epi-LXA4 or 17-epi-RvD1 at the peak of inflammation accelerates clearance of bacteria, blunts PMN accumulation, and promotes PMN apoptosis and efferocytosis, thereby facilitating resolution of E. coli-evoked lung injury. Collectively, these results uncover a TLR9-mediated endogenous mechanism that impairs PMN phagocytosis and prolongs inflammation, and demonstrate both endogenous and therapeutic potential for 15-epi-LXA4 and 17-epi-RvD1 to restore impaired bacterial clearance and facilitate resolution of acute lung inflammation.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Escherichia coli Infections/immunology , Neutrophils/immunology , Phagocytosis/immunology , Pneumonia/immunology , Toll-Like Receptor 9/metabolism , Adult , Aged , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Cells, Cultured , CpG Islands/immunology , DNA, Bacterial/immunology , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Healthy Volunteers , Humans , Lipoxins/pharmacology , Lipoxins/therapeutic use , Lung/microbiology , Lung/pathology , Male , Mice , Middle Aged , Neutrophils/metabolism , Phagocytosis/drug effects , Pneumonia/drug therapy , Pneumonia/microbiology , Pneumonia/pathology , Primary Cell Culture , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/immunology , Receptors, Lipoxin/metabolism
2.
Cytokine Growth Factor Rev ; 50: 29-42, 2019 12.
Article En | MEDLINE | ID: mdl-31718982

Expression of the acute phase protein serum amyloid A (SAA) is dependent on the release of the pro-inflammatory cytokines IL-1, IL-6 and TNF-α during infection and inflammation. Hepatitis C virus (HCV) upregulates SAA-inducing cytokines. In line with this, a segment of chronically infected individuals display increased circulating levels of SAA. SAA has even been proposed to be a potential biomarker to evaluate treatment efficiency and the course of disease. SAA possesses antiviral activity against HCV via direct interaction with the viral particle, but might also divert infectivity through its function as an apolipoprotein. On the other hand, SAA shares inflammatory and angiogenic activity with chemotactic cytokines by activating the G protein-coupled receptor, formyl peptide receptor 2. These latter properties might promote chronic inflammation and hepatic injury. Indeed, up to 80 % of infected individuals develop chronic disease because they cannot completely clear the infection, due to diversion of the immune response. In this review, we summarize the interconnection between SAA and cytokines in the context of HCV infection and highlight the dual role SAA could play in this disease. Nevertheless, more research is needed to establish whether the balance between those opposing activities can be tilted in favor of the host defense.


Cytokines/immunology , Hepatitis C/immunology , Hepatitis C/physiopathology , Serum Amyloid A Protein/immunology , Animals , Cytokines/genetics , Hepacivirus , Humans , Inflammation , Liver/immunology , Liver/pathology , Mice , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , Serum Amyloid A Protein/analysis
3.
Front Immunol ; 10: 938, 2019.
Article En | MEDLINE | ID: mdl-31114582

Annexin-A1 (ANXA1) was first discovered in the early 1980's as a protein, which mediates (some of the) anti-inflammatory effects of glucocorticoids. Subsequently, the role of ANXA1 in inflammation has been extensively studied. The biology of ANXA1 is complex and it has many different roles in both health and disease. Its effects as a potent endogenous anti-inflammatory mediator are well-described in both acute and chronic inflammation and its role in activating the pro-resolution phase receptor, FPR2, has been described and is now being exploited for therapeutic benefit. In the present mini review, we will endeavor to give an overview of ANXA1 biology in relation to inflammation and functions that mediate pro-resolution that are independent of glucocorticoid induction. We will focus on the role of ANXA1 in diseases with a large inflammatory component focusing on diabetes and microvascular disease. Finally, we will explore the possibility of exploiting ANXA1 as a novel therapeutic target in diabetes and the treatment of microvascular disease.


Annexin A1/immunology , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Vascular Diseases/immunology , Addison Disease/drug therapy , Addison Disease/immunology , Addison Disease/pathology , Animals , Cushing Syndrome/drug therapy , Cushing Syndrome/immunology , Cushing Syndrome/pathology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/immunology , Diabetes Mellitus/pathology , Glucocorticoids/immunology , Glucocorticoids/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Vascular Diseases/drug therapy , Vascular Diseases/pathology
4.
Parasit Vectors ; 12(1): 275, 2019 May 29.
Article En | MEDLINE | ID: mdl-31142352

BACKGROUND: Neutrophils play an immunomodulatory role through the release of neutrophil extracellular traps (NETs). NETs are released in response to Leishmania infection, but the mechanism of NET extrusion has not been elucidated. The lipoxin A4 receptor on neutrophils is crucial for the inflammatory response and immune regulation of many diseases, including Leishmania infection. Therefore, in the present study, we tried to explore whether Leishmania infantum promastigotes stimulate neutrophil activation and NET release via activating the lipoxin A4 receptor. RESULTS: Leishmania infantum promastigotes stimulated neutrophil activity, but blocking of the lipoxin A4 receptor with its antagonist Boc prior to L. infantum stimulation abrogated these effects. Neutrophils showed citrullinated histone H3 expression and simultaneous NET extrusion on L. infantum stimulation, but a decline in both was observed on blocking of the lipoxin A4 receptor. Moreover, differentiated HL-60 cells with lipoxin A4 receptor silencing showed a decrease in citrullinated histone H3 expression as compared to the unsilenced HL-60 samples on stimulation with promastigotes. CONCLUSIONS: Leishmania infantum promastigotes altered the characteristics of neutrophils and induced NET extrusion by activating the lipoxin A4 receptor. The lipoxin A4 receptor may have potential as a therapeutic target in relation to NET extrusion in the treatment of leishmaniasis, but its mechanisms of action need to be explored in more depth.


Extracellular Traps/immunology , Leishmania infantum/immunology , Neutrophils/immunology , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Animals , Citrullination , Female , Gene Silencing , HL-60 Cells , Histones/metabolism , Humans , Lipoxins/pharmacology , Mice , Mice, Inbred BALB C , Neutrophils/parasitology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors
5.
J Immunol ; 202(9): 2710-2719, 2019 05 01.
Article En | MEDLINE | ID: mdl-30902901

Similar to bacteria, synthesis of mitochondrial DNA-encoded proteins requires an N-formylated methionine to initiate translation. Thus, the N-formylated methionine peptides originating from mitochondria should be recognized as danger signals. To date, only one such peptide, denoted as mitocryptide-2 (MCT-2), originating from the N-terminal of the mitochondrial cytochrome b, has been isolated from mammalian tissues. Human neutrophils express FPR1 and FPR2 that detect formyl peptides, and the precise structural determinants for receptor recognition remain to be elucidated. MCT-2 is known to activate neutrophils through FPR2 but not FPR1. The aim of this study was to elucidate the structural determinants of importance for receptor preference and human neutrophil activation in MCT-2 by generating a series of MCT-2 variants. We show that there is an absolute requirement for the N-formyl group and the side chain of Met1 at position 1 of MCT-2 but also the C terminus is of importance for MCT-2 activity. We also uncovered individual side chains that positively contribute to MCT-2 activity as well as those suppressed in the response. The MCT-2 peptide and its two polymorphic variants ([Thr7]MCT-2 and [Ser8]MCT-2) all activated neutrophils, but MCT-2 containing Ile7 and Asn8 was the most potent. We also show that some peptide variants displayed a biased FPR2-signaling property related to NADPH oxidase activation and ß-arrestin recruitment, respectively. In conclusion, we disclose several critical elements in MCT-2 that are required for neutrophil activation and disclose structural insights into how FPR2 recognition of this mitochondrial DNA-derived peptide may increase our understanding of the role of FPR2 in aseptic inflammation.


Cytochromes b/immunology , DNA, Mitochondrial/immunology , Mitochondrial Proteins/immunology , Neutrophils/immunology , Peptides/immunology , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Cytochromes b/chemistry , Female , Humans , Male , Mitochondrial Proteins/chemistry , Peptides/chemistry , Receptors, Formyl Peptide/chemistry , Receptors, Lipoxin/chemistry
6.
Blood ; 131(4): 439-449, 2018 01 25.
Article En | MEDLINE | ID: mdl-29371208

A natural leukocyte chemoattractant was isolated from bovine serum by an established 4-step purification procedure. Based on its relative molecular mass of 7287 and NH2-terminal sequence, the protein was identified as a carboxy-terminal peptide of the acute phase protein serum amyloid A1 (SAA1). This SAA1(46-112) fragment and its human equivalent SAA1(47-104) were chemically synthesized. Unlike intact SAA1α, these SAA fragments failed to directly chemoattract neutrophils and monocytes, to induce chemokines, and to stimulate downstream extracellular signal-regulated kinase signaling in monocytes. However, the SAA fragments potently synergized with CCL3 to induce monocyte migration and with CXCL8 to stimulate neutrophil shape changes and chemotaxis. Unlike intact SAA1α, SAA1(46-112) did not induce CXCL6 ex vivo but provoked a cooperative intraperitoneal neutrophil recruitment in mice when coinjected with CXCL6 into the peritoneal cavity. Moreover, SAA1(47-104) desensitized the synergy between intact SAA1α and CXCL8 in neutrophil chemotaxis, suggesting that this peptide binds formyl peptide receptor 2 (FPR2). This was evidenced by a complete blockade of synergy between the COOH-terminal SAA1 fragments and CXCL8 or CCL3 in neutrophil and monocyte chemotaxis, respectively, by the FPR2 antagonist WRW4 Thus, SAA1 is degraded into fragments lacking chemokine-inducing capacity, while keeping synergy with cytokine-induced chemokines to sustain limited inflammation.


Chemokine CCL3/immunology , Chemokines/immunology , Interleukin-8/immunology , Leukocytes/drug effects , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Serum Amyloid A Protein/chemistry , Serum Amyloid A Protein/pharmacology , Animals , Cattle , Chemotaxis/drug effects , Female , Humans , Leukocytes/immunology , Mice , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Serum Amyloid A Protein/chemical synthesis
7.
FASEB J ; 32(1): 26-36, 2018 01.
Article En | MEDLINE | ID: mdl-28855276

Leukocytes express formyl-peptide receptors (FPRs), which sense microbe-associated molecular pattern (MAMP) molecules, leading to leukocyte chemotaxis and activation. We recently demonstrated that phenol-soluble modulin (PSM) peptides from highly pathogenic Staphylococcus aureus are efficient ligands for the human FPR2. How PSM detection by FPR2 impacts on the course of S. aureus infections has remained unknown. We characterized the specificity of mouse FPR2 (mFpr2) using a receptor-transfected cell line, homeobox b8 (Hoxb8), and primary neutrophils isolated from wild-type (WT) or mFpr2-/- mice. The influx of leukocytes into the peritoneum of WT and mFpr2-/- mice was analyzed. We demonstrate that mFpr2 is specifically activated by PSMs in mice, and they represent the first secreted pathogen-derived ligands for the mFpr2. Intraperitoneal infection with S. aureus led to lower numbers of immigrated leukocytes in mFpr2-/- compared with WT mice at 3 h after infection, and this difference was not observed when mice were infected with an S. aureus PSM mutant. Our data support the hypothesis that the mFpr2 is the functional homolog of the human FPR2 and that a mouse infection model represents a suitable model for analyzing the role of PSMs during infection. PSM recognition by mFpr2 shapes leukocyte influx in local infections, the typical infections caused by S. aureus-Weiss, E., Hanzelmann, D., Fehlhaber, B., Klos, A., von Loewenich, F. D., Liese, J., Peschel, A., Kretschmer, D. Formyl-peptide receptor 2 governs leukocyte influx in local Staphylococcus aureus infections.


Leukocytes/immunology , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Staphylococcal Infections/immunology , Animals , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Calcium Signaling/immunology , Cell Degranulation/immunology , Cell Line , Cell Movement/immunology , Disease Models, Animal , Female , Genes, Bacterial , Homeodomain Proteins/immunology , Humans , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neutrophils/immunology , Receptors, Formyl Peptide/deficiency , Receptors, Formyl Peptide/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology
8.
Sci Rep ; 7(1): 6718, 2017 07 27.
Article En | MEDLINE | ID: mdl-28751674

Formyl peptide receptor 1 (FPR1) mediates bacterial and mitochondrial N-formyl peptides-induced neutrophil activation. Therefore, FPR1 is an important therapeutic target for drugs to treat septic or sterile inflammatory diseases. Honokiol, a major bioactive compound of Magnoliaceae plants, possesses several anti-inflammatory activities. Here, we show that honokiol exhibits an inhibitory effect on FPR1 binding in human neutrophils. Honokiol inhibited superoxide anion generation, reactive oxygen species formation, and elastase release in bacterial or mitochondrial N-formyl peptides (FPR1 agonists)-activated human neutrophils. Adhesion of FPR1-induced human neutrophils to cerebral endothelial cells was also reduced by honokiol. The receptor-binding results revealed that honokiol repressed FPR1-specific ligand N-formyl-Nle-Leu-Phe-Nle-Tyr-Lys-fluorescein binding to FPR1 in human neutrophils, neutrophil-like THP-1 cells, and hFPR1-transfected HEK293 cells. However, honokiol did not inhibit FPR2-specific ligand binding to FPR2 in human neutrophils. Furthermore, honokiol inhibited FPR1 agonist-induced calcium mobilization as well as phosphorylation of p38 MAPK, ERK, and JNK in human neutrophils. In conclusion, our data demonstrate that honokiol may have therapeutic potential for treating FPR1-mediated inflammatory diseases.


Anti-Inflammatory Agents/pharmacology , Biphenyl Compounds/pharmacology , Endothelial Cells/drug effects , Lignans/pharmacology , Neutrophils/drug effects , Oligopeptides/antagonists & inhibitors , Receptors, Formyl Peptide/genetics , Animals , Anti-Inflammatory Agents/isolation & purification , Biphenyl Compounds/isolation & purification , Brain/cytology , Brain/metabolism , Cell Adhesion/drug effects , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Lignans/isolation & purification , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , Magnolia/chemistry , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Neutrophil Activation/drug effects , Neutrophils/cytology , Neutrophils/immunology , Oligopeptides/pharmacology , Plant Extracts/chemistry , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , THP-1 Cells , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
9.
Stroke ; 47(2): 490-7, 2016 Feb.
Article En | MEDLINE | ID: mdl-26732571

BACKGROUND AND PURPOSE: Lipoxin A4 (LXA4) has been reported to reduce inflammation in several neurological injury models. We studied the effects of LXA4 on neuroinflammation after subarachnoid hemorrhage (SAH) in a rat model. METHODS: Two hundred and thirty-eight Sprague-Dawley male rats, weight 280-320 g, were used. Exogenous LXA4 (0.3 and 1.0 nmol) were injected intracerebroventricularly at 1.5 hours after SAH. Neurological scores, brain water content, and blood-brain barrier were evaluated at 24 hours after SAH; Morris water maze and T-maze tests were examined at 21 days after SAH. The expression of endogenous LXA4 and its receptor formyl peptide receptor 2 (FPR2), as well as p38, interleukin-1ß, and interleukin-6 were studied either by ELISA or by Western blots. Neutrophil infiltration was observed by myeloperoxidase staining. FPR2 siRNA was used to knock down LXA4 receptor. RESULTS: The expression of endogenous LXA4 decreased, and the expression of FPR2 increased after SAH. Exogenous LXA4 decreased brain water content, reduced Evans blue extravasation, and improved neurological functions and improved the learning and memory ability after SAH. LXA4 reduced neutrophil infiltration and phosphorylation of p38, interleukin-1ß, and interleukin-6. These effects of LXA4 were abolished by FPR2 siRNA. CONCLUSIONS: Exogenous LXA4 inhibited inflammation by activating FPR2 and inhibiting p38 after SAH. LXA4 may serve as an alternative treatment to relieve early brain injury after SAH.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Behavior, Animal/drug effects , Lipoxins/pharmacology , Receptors, Lipoxin/drug effects , Subarachnoid Hemorrhage/immunology , p38 Mitogen-Activated Protein Kinases/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blotting, Western , Brain Edema/immunology , Brain Edema/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Inflammation , Injections, Intraventricular , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lipoxins/immunology , Lipoxins/metabolism , Male , Memory/drug effects , Neuropsychological Tests , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Rats , Rats, Sprague-Dawley , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Spatial Learning/drug effects , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Leukoc Biol ; 98(6): 1049-60, 2015 Dec.
Article En | MEDLINE | ID: mdl-26297794

Cell migration depends on the ability of leukocytes to sense an external gradient of chemotactic proteins produced during inflammation. These proteins include chemokines, complement factors, and some acute phase proteins, such as serum amyloid A. Serum amyloid A chemoattracts neutrophils, monocytes, and T lymphocytes via its G protein-coupled receptor formyl peptide receptor 2. We demonstrate that serum amyloid A1α more potently chemoattracts neutrophils in vivo than in vitro. In contrast to CD14(+) monocytes, no rapid (within 2 h) induction of interleukin-8/CXC chemokine ligand 8 or macrophage-inflammatory protein-1α/CC chemokine ligand 3 was observed in purified human neutrophils after stimulation of the cells with serum amyloid A1α or lipopolysaccharide. Moreover, interleukin-8/CXC chemokine ligand 8 induction in monocytes by serum amyloid A1α was mediated by toll-like receptor 2 and was inhibited by association of serum amyloid A1α with high density lipoprotein. This indicates that the potent chemotactic response of neutrophils toward intraperitoneally injected serum amyloid A1α is indirectly enhanced by rapid induction of chemokines in peritoneal cells, synergizing in a paracrine manner with serum amyloid A1α. We observed direct synergy between IL-8/CXC chemokine ligand 8 and serum amyloid A1α, but not lipopolysaccharide, in chemotaxis and shape change assays with neutrophils. Furthermore, the selective CXC chemokine receptor 2 and formyl peptide receptor 2 antagonists, SB225002 and WRW4, respectively, blocked the synergy between IL-8/CXC chemokine ligand 8 and serum amyloid A1α in neutrophil chemotaxis in vitro, indicating that for synergy their corresponding G protein-coupled receptors are required. Additionally, SB225002 significantly inhibited serum amyloid A1α-mediated peritoneal neutrophil influx. Taken together, endogenous (e.g., IL-1ß) and exogenous (e.g., lipopolysaccharide) inflammatory mediators induce primary chemoattractants such as serum amyloid A that synergize in an autocrine (monocyte) or a paracrine (neutrophil) fashion with secondary chemokines induced in stromal cells.


Chemotaxis, Leukocyte/immunology , Interleukin-8/immunology , Neutrophils/immunology , Paracrine Communication/immunology , Receptors, Formyl Peptide/immunology , Receptors, Interleukin-8B/immunology , Receptors, Lipoxin/immunology , Serum Amyloid A Protein/immunology , Toll-Like Receptor 2/immunology , Chemotaxis, Leukocyte/drug effects , Female , Humans , Male , Paracrine Communication/drug effects , Phenylurea Compounds/pharmacology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/antagonists & inhibitors
11.
Eur J Immunol ; 44(2): 357-69, 2014 Feb.
Article En | MEDLINE | ID: mdl-24166736

Specialized proresolving mediators are endogenous bioactive lipid molecules that play a fundamental role in the regulation of inflammation and its resolution. Lipoxins and other specialized proresolving mediators have been identified in important immunological tissues including bone marrow, spleen, and blood. Lipoxins regulate functions of the innate immune system including the promotion of monocyte recruitment and increase macrophage phagocytosis of apoptotic neutrophils. A major knowledge gap is whether lipoxins influence adaptive immune cells. Here, we analyzed the actions of lipoxin A4 (LXA4) and its receptor ALX/FPR2 on human and mouse B cells. LXA4 decreased IgM and IgG production on activated human B cells through ALX/FPR2-dependent signaling, which downregulated NF-κB p65 nuclear translocation. LXA4 also inhibited human memory B-cell antibody production and proliferation, but not naïve B-cell function. Lastly, LXA4 decreased antigen-specific antibody production in an OVA immunization mouse model. To our knowledge, this is the first description of the actions of lipoxins on human B cells, demonstrating a link between resolution signals and adaptive immunity. Regulating antibody production is crucial to prevent unwanted inflammation. Harnessing the ability of lipoxins to decrease memory B-cell antibody production can be beneficial to threat inflammatory and autoimmune disorders.


Adaptive Immunity/immunology , Adaptor Proteins, Signal Transducing/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Lipoxins/immunology , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Animals , Antibodies/immunology , Cell Proliferation , Cells, Cultured , Down-Regulation/immunology , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Protein Transport/immunology , Signal Transduction/immunology , Up-Regulation/immunology
12.
J Immunol ; 190(12): 6478-87, 2013 Jun 15.
Article En | MEDLINE | ID: mdl-23686496

Endogenous mechanisms regulating the host response during inflammation resolution are critical in ensuring disposal of noxious stimuli and return to homeostasis. In this article, we engineered novel Annexin A1 (AnxA1)-based peptides, AnxA1(2-50), that displayed specific binding to the AnxA1 receptor (formyl peptide receptor 2/Lipoxin A4 receptor [FPR2/ALX]; IC50 ∼4 nM). Intravenous administration of AnxA1(2-50) markedly reduced (>60%) leukocyte adhesion to postcapillary venules in wild type and Fpr1(-/-), but not Fpr2/Alx(-/-), mice. Generation of a metabolically stable form of this peptide (CR-AnxA1(2-50)), engineered by substituting a cleavage site shared by human proteinase 3 and neutrophil elastase, yielded an agonist that was resistant to neutrophil-mediated cleavage and displayed enhanced proresolving actions: accelerated resolution of self-limited inflammation and enhanced macrophage efferocytosis after sterile injury, when compared with AnxA1(2-50). These actions were retained with human primary leukocytes where CR-AnxA1(2-50) decreased neutrophil-endothelial interactions (∼25-45%), and stimulated neutrophil apoptosis and macrophage efferocytosis (∼45%). In murine cardiac ischemia/reperfusion injury, CR-AnxA1(2-50) elicited tissue-protective actions reducing infarct size (∼60%) and incidence of 24-h death. These results identify AnxA1(2-50) and CR-AnxA1(2-50) as FPR2/ALX agonists that harness the proresolving actions of AnxA1, and thus may represent therapeutic tools for treatment of inflammatory conditions.


Annexin A1/immunology , Anti-Inflammatory Agents/immunology , Inflammation/immunology , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/agonists , Receptors, Lipoxin/immunology , Animals , Annexin A1/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Neutrophils/metabolism , Peptides/immunology , Phagocytosis/immunology
13.
J Innate Immun ; 4(2): 201-12, 2012.
Article En | MEDLINE | ID: mdl-22067547

The Agr quorum-sensing system represents the master regulator for staphylococcal virulence factors and is known to have a strong impact on the release of pathogen-associated molecular pattern (PAMP) molecules. Among the various staphylococcal PAMPs, phenol-soluble modulin (PSM) peptides have attracted increasing interest because they are crucial for staphylococcal virulence and have neutrophil-recruiting properties. The latter depend on recognition of PSMs by the neutrophil formyl peptide receptor 2 (FPR2/ALX), for which PSMs are highly efficient agonists. We demonstrate that Agr inactivation in Staphylococcus aureus or S. epidermidis leads to strongly reduced neutrophil responses, which is in agreement with the previously reported strict control of PSM expression by Agr. Agr had a distinct and profound impact on activation of FPR2/ALX but not of the related FPR1 receptor that senses bacterial formylated peptides. S. epidermidis PSMs had similar FPR2/ALX-activating properties but differed in their dependence on N-terminal formylation compared to S. aureus PSMs. Moreover, S. aureus and S. epidermidis PSMs upregulated the neutrophil complement receptor CD11b via FPR2/ALX stimulation in an Agr-dependent fashion. Hence, Agr controls the capacity of staphylococcal pathogens to activate FPR2/ALX-dependent neutrophil responses, underscoring the crucial role of FPR2/ALX and PSMs in staphylococcus-host interaction.


Bacterial Proteins/immunology , Neutrophils/immunology , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Staphylococcus aureus/immunology , Staphylococcus epidermidis/immunology , Trans-Activators/immunology , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Bacterial Proteins/metabolism , Cell Line , Host-Pathogen Interactions/immunology , Humans , Neutrophils/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Staphylococcus epidermidis/metabolism , Staphylococcus epidermidis/pathogenicity , Trans-Activators/metabolism , Virulence
14.
Mediators Inflamm ; 2011: 745340, 2011.
Article En | MEDLINE | ID: mdl-21765620

Fibroblasts are important to host defence and immunity, can also as initiators of inflammation as well. As the endogenous "braking signal", Lipoxins can regulate anti-inflammation and the resolution of inflammation. We investigated the effect of lipoxinA(4) on the expression of cyclooxygenase-2 in lipopolysaccharide-stimulated lung fibroblasts. We demonstrated that the expression of cyclooxygenase-2 protein was significantly increased and peaked initially at 6 hours, with a second increase, with maximal levels occurring 24 hours after lipopolysaccharide challenge. ProstaglandinE(2) levels also peaked at 6 hours, and prostaglandinD(2) levels were increased at both 6 and 24 hours. Exogenous lipoxinA(4) inhibited the first peak of cyclooxygenase-2 expression as well as the production of prostaglandinE(2) induced by lipopolysaccharide in a dose-dependent manner. In contrast, exogenous lipoxinA(4) increased the second peak of cyclooxygenase-2 expression as well as the production of prostaglandinD(2) induced by lipopolysaccharide in a dose-dependent manner. LipoxinA(4) receptor mRNA expression was markedly stimulated by lipopolysaccharide but inhibited by lipoxinA(4). We present evidence for a novel biphasic role of lipoxinA(4) on the expression of cyclooxygenase-2 in lipopolysaccharide-stimulated lung fibroblasts, whereby LXA(4) has an anti-inflammatory and proresolving activity in lung fibroblasts following LPS stimulation.


Cyclooxygenase 2/immunology , Fibroblasts/immunology , Lipopolysaccharides/pharmacology , Lipoxins/immunology , Lung/cytology , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Lipoxins/metabolism , Lipoxins/pharmacology , Prostaglandin D2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Lipoxin/immunology , Receptors, Lipoxin/metabolism , Up-Regulation/immunology
15.
J Cell Biochem ; 112(8): 2097-105, 2011 Aug.
Article En | MEDLINE | ID: mdl-21465531

Serum amyloid A (SAA) is regarded as an important acute phase protein in coronary artery diseases. However, its involvement in the immune response of atherosclerosis is poorly understood. The present study was designed to investigate the influence of SAA on the secretion of long pentraxin 3 (PTX3), a key component of innate immunity, in human aortic endothelial cells (HAECs). Our study revealed that recombinant SAA up-regulated PTX3 production in a remarkable dose- and time-dependent manner and the activation of formyl peptide receptor-like 1 (FPRL1) was crucial for SAA-induced expression of PTX3 in HAECs. Meanwhile, SAA-induced PTX3 production could be significantly down-regulated by using the specific siRNA sequences for Jun N-terminal kinases (JNK). Furthermore, the activation of activator protein-1 (AP-1) was necessary for the up-regulation of PTX3 expression. We also found that the activation of nuclear factor-kappa B (NF-κB) played an important role in this process. Our findings demonstrate that SAA up-regulates PTX3 production via FPRL1 significantly, and thus, contributes to the inflammatory pathogenesis of atherosclerosis.


Aorta/metabolism , C-Reactive Protein/biosynthesis , Endothelial Cells/metabolism , Immunity, Innate/drug effects , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Serum Amyloid A Protein/pharmacology , Serum Amyloid P-Component/biosynthesis , Signal Transduction/drug effects , Aorta/immunology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , C-Reactive Protein/genetics , C-Reactive Protein/immunology , Cell Line , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Immunity, Innate/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , MAP Kinase Kinase 4/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/immunology , Serum Amyloid A Protein/metabolism , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Transcription Factor AP-1/metabolism
16.
J Immunol ; 186(8): 4905-14, 2011 Apr 15.
Article En | MEDLINE | ID: mdl-21398608

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.


Microvessels/immunology , Neutrophils/immunology , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Animals , Annexin A1/genetics , Annexin A1/immunology , Annexin A1/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/prevention & control , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lipoxins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/pathology , Neutrophils/drug effects , Neutrophils/metabolism , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Protein Transport/drug effects , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Time Factors
17.
J Acquir Immune Defic Syndr ; 56(1): 9-15, 2011 Jan 01.
Article En | MEDLINE | ID: mdl-20921899

BACKGROUND: Coreceptor switching from CCR5 to CXCR4 is less common in subtype C HIV-1 infection than in subtype B for reasons that are unclear. We have examined sequential virus samples from a subtype C-infected child who had evidence of coreceptor switching. METHODS: To examine HIV-1 envelope evolution towards CXCR4 usage, env sequences were correlated with phenotypic characteristics determined by entry assays, as well as the ability to use alternative coreceptors such as FPRL1, CCR3, CCR8 and others. The value of a phenotype predictor based on V3 sequences was also assessed. RESULTS: Ninety-three sequences revealed 3 distinct coexistent virus lineages and only some members of one lineage evolved to use CXCR4. These lineages also had diverse alternative coreceptor patterns including the ability to use FPRL1, CCR3, CCR8, APJ, CMKLR1, RDC-1, CXCR6, CCR1, GPCR1, GPR15 and CCR6. Coreceptor switching was associated with extensive and rapid sequence divergence in the V1/V2 region in addition to V3 changes. Furthermore, interlineage recombination within the C2 region resulted in low predictability of a V3 sequence-based phenotype algorithm, and highlighted the importance of V1/V2 and V3 sequences in coreceptor usage. CONCLUSION: These results suggest that the evolution to coreceptor switching in subtype C infection requires more mutations than other subtypes, and this contributes to the reduced incidence of R5X4 viruses.


HIV-1/genetics , Receptors, CCR5/genetics , Receptors, CXCR4/genetics , Child , Cloning, Molecular , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Humans , Molecular Sequence Data , Phenotype , Phylogeny , Receptors, CCR5/immunology , Receptors, CXCR4/immunology , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , Recombination, Genetic/genetics , env Gene Products, Human Immunodeficiency Virus/genetics
18.
FEBS Lett ; 584(18): 4102-8, 2010 Sep 24.
Article En | MEDLINE | ID: mdl-20804753

We synthesized and investigated the effect of formyl peptide receptor 2 (FPR2)-derived pepducins in human monocytes. The FPR2-based cell-penetrating lipopeptide, "pepducin" (F2pal-16), stimulated intracellular calcium increase in human monocytes via pertussis toxin (PTX)-sensitive G-protein and phospholipase C (PLC) activity. From a functional aspect, we showed that F2pal-16 stimulated monocyte chemotaxis. F2pal-16 also stimulated the generation of superoxide anion in human monocytes. Moreover, F2pal-16 dramatically increased the production of several kinds of pro-inflammatory cytokines (CXCL8, CCL2, IL-1ß and TNF-α) in human monocytes via NF-κB activation. Since FPR2 plays an important role in immune responses, F2pal-16 can serve as a useful reagent for the study of FPR2-mediated immune modulation.


Chemotaxis/drug effects , Lipopeptides/pharmacology , Monocytes/drug effects , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Amino Acid Sequence , Calcium/metabolism , Cells, Cultured , Chemokine CCL2/biosynthesis , Chemotaxis/immunology , Cytokines/biosynthesis , Humans , Lipopeptides/chemical synthesis , Molecular Sequence Data , Monocytes/immunology , NF-kappa B/metabolism , Superoxides/metabolism
19.
Free Radic Biol Med ; 48(2): 207-15, 2010 Jan 15.
Article En | MEDLINE | ID: mdl-19854263

Antimicrobial defense by neutrophils implicates the production of reactive oxygen species. Neutrophil responses can be modulated by agonists such as bacterial peptides and proinflammatory factors that modulate neutrophil activity and survival. We evaluated the production of superoxide anions (O(2)(-)) in response to fMLF by normal human neutrophils after 3 days of preincubation with GM-CSF + IL-4 + TNF-alpha (survival medium). After 3 days of incubation in survival medium, long-lived neutrophils produced up to six times more O(2)(-) relative to control neutrophils in response to fMLF and WKYMVM. This augmented response to fMLF was preferentially linked to formyl peptide receptor (FPR), whereas the response to WKYMVM was dependent on formyl peptide receptor-like 1 (FPRL-1). Real-time RT-PCR revealed a diminution of FPR and FPRL-1 expression in neutrophils incubated in survival medium. fMLF-induced overproduction of O(2)(-) by long-lived neutrophils was independent of intracellular calcium mobilization. The protein tyrosine phosphorylation profile of long-lived neutrophils was modified with respect to control cells. Pharmacological inhibitors of intracellular signals indicated that mechanisms of the excessive fMLF-induced production of O(2)(-) by long-lived neutrophils implicated the protein kinase C (PKC) pathway, preferentially the PKC-delta isoform, whose protein was augmented in these cells. Thus, long-term cytokine exposure modifies molecular pathways and functional characteristics of the neutrophil.


Cytokines/immunology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/metabolism , Protein Kinase C-delta/metabolism , Superoxides/metabolism , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Down-Regulation/immunology , Humans , Immunity, Innate , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Oligopeptides/pharmacology , Protein Kinase C-delta/immunology , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , Receptors, Lipoxin/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Superoxides/immunology
20.
Immunobiology ; 215(1): 38-52, 2010.
Article En | MEDLINE | ID: mdl-19250699

Exocytosis of neutrophil granules is a major event that converts circulating neutrophils into fully activated cells capable of chemotaxis, phagocytosis and destruction of pathogens. The PLB-985 cell line is a suitable neutrophilic cellular model which is utilised to study the different functional responses of neutrophils. In this study, we characterised the differentiation of PLB-985 cells toward the granulocytic pathway, using three different inducing agents: dbcAMP, DMSO and DMF. The differentiation efficiency was monitored by observation of cell morphology with electron microscopy, and by analysis of the expression of receptors such as FPRL1 and FcgammaRIIA, the distribution or release of granule markers, phagocytic capacity, as well as measurement of fMLF-induced calcium fluxes. Exocytosis and phagocytosis in differentiated cells were weaker as compared to neutrophils. fMLF stimulated primary granule exocytosis in cells differentiated with dbcAMP, DMSO and DMF, whereas the release of the contents of tertiary granules, as well as that of secretory vesicles, was only observed in dbcAMP-differentiated cells. DMSO-differentiated cells exhibited the highest phagocytic capacity. Altogether our results reinforce the fact that depending on the differentiating agent used, PLB-985 cells represent a useful model to study neutrophil functions and to bypass difficulties inherent to these primary cells.


Cell Degranulation/immunology , Cell Differentiation/immunology , Cell Line , Neutrophils/cytology , Neutrophils/metabolism , Phagocytosis/immunology , Bucladesine/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/immunology , Cell Degranulation/drug effects , Cell Differentiation/drug effects , Dimethyl Sulfoxide/pharmacology , Dimethylformamide/pharmacology , Granulocytes/cytology , Humans , Microscopy, Electron , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Phagocytosis/drug effects , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/metabolism , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, IgG/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , Receptors, Lipoxin/metabolism
...