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










Publication year range
1.
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
2.
Biochim Biophys Acta ; 1833(8): 1914-23, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23562731

ABSTRACT

Lipidated peptides (pepducins) can activate certain G-protein coupled receptors (GPCRs) through a unique allosteric modulation mechanism involving cytosolic receptor domains. Pepducins with the amino acid sequence of the third intracellular loop of the neutrophil formyl peptide receptors (FPRs) as a common denominator were N-terminally conjugated with palmitic acid. F2Pal16, containing the 16 amino acids present in the third intracellular loop of FPR2, induced superoxide production in human neutrophils and the activity was sensitive to FPR2 antagonists. Cells over-expressing FPR2 were similarly responsive and responded with a transient increase in cytosolic calcium. No such effects were observed with the corresponding FPR1 pepducin. The peptide alone, lacking palmitic acid, did not activate neutrophils. A ten amino acid long pepducin F2Pal10, that was a more potent neutrophil activator than F2Pal16, was used for amino acid substitution studies. The sequences of FPR1 and FPR2 in the third intracellular loop differ by only two amino acids, and a pepducin with the FPR2-specific K231 replaced by the FPR1-specific Q231 lost all activity. The active F2Pal10 pepducin also triggered a response in cells expressing a mutated FPR2 with the third intracellular loop identical to that of FPR1. The data presented suggest that the same signaling pathways are activated when the signaling cascade is initiated by a classical receptor agonist (outside-in signaling) and when signaling starts on the cytosolic side of the membrane by a pepducin (inside-in signaling). A fundamental difference is also disclosed between the two neutrophil FPRs regarding their sensitivities to third intracellular loop pepducins.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cytosol/metabolism , HL-60 Cells , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Molecular Sequence Data , Neutrophils/drug effects , Neutrophils/metabolism , Protein Structure, Tertiary , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Signal Transduction/drug effects , Structure-Activity Relationship
3.
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
4.
PLoS One ; 7(1): e30488, 2012.
Article in English | MEDLINE | ID: mdl-22299042

ABSTRACT

Pseudomonas aeruginosa type III secretion apparatus exports and translocates four exotoxins into the cytoplasm of the host cell. The translocation requires two hydrophobic bacterial proteins, PopB and PopD, that are found associated with host cell membranes following infection. In this work we examined the influence of host cell elements on exotoxin translocation efficiency. We developed a quantitative flow cytometry based assay of translocation that used protein fusions between either ExoS or ExoY and the ß-lactamase reporter enzyme. In parallel, association of translocon proteins with host plasma membranes was evaluated by immunodetection of PopB/D following sucrose gradient fractionation of membranes. A pro-myelocytic cell line (HL-60) and a pro-monocytic cell line (U937) were found resistant to toxin injection even though PopB/D associated with host cell plasma membranes. Differentiation of these cells to either macrophage- or neutrophil-like cell lines resulted in injection-sensitive phenotype without significantly changing the level of membrane-inserted translocon proteins. As previous in vitro studies have indicated that the lysis of liposomes by PopB and PopD requires both cholesterol and phosphatidyl-serine, we first examined the role of cholesterol in translocation efficiency. Treatment of sensitive HL-60 cells with methyl-ß-cyclodextrine, a cholesterol-depleting agent, resulted in a diminished injection of ExoS-Bla. Moreover, the PopB translocator was found in the membrane fraction, obtained from sucrose-gradient purifications, containing the lipid-raft marker flotillin. Examination of components of signalling pathways influencing the toxin injection was further assayed through a pharmacological approach. A systematic detection of translocon proteins within host membranes showed that, in addition to membrane composition, some general signalling pathways involved in actin polymerization may be critical for the formation of a functional pore. In conclusion, we provide new insights in regulation of translocation process and suggest possible cross-talks between eukaryotic cell and the pathogen at the level of exotoxin translocation.


Subject(s)
Bacterial Translocation , Exotoxins/administration & dosage , Exotoxins/genetics , Host-Pathogen Interactions/genetics , Pseudomonas aeruginosa/genetics , ADP Ribose Transferases/administration & dosage , ADP Ribose Transferases/genetics , ADP Ribose Transferases/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Bacterial Toxins/pharmacology , Bacterial Translocation/genetics , Bacterial Translocation/physiology , Cell Membrane/metabolism , Exotoxins/metabolism , Exotoxins/pharmacology , Genes, Reporter , HL-60 Cells , Host-Pathogen Interactions/drug effects , Humans , Immunity, Innate/genetics , Injections , Protein Transport , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/physiology , U937 Cells , beta-Lactamases/genetics , beta-Lactamases/metabolism
5.
J Biol Chem ; 286(30): 26718-31, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21543323

ABSTRACT

Among human N-formyl peptide chemoattractant receptors, FPR2/ALX and FPR3 share the highest degree of amino acid identity (83%), and trigger similar cell responses upon ligand binding. Although FPR2/ALX is a promiscuous receptor, FPR3 has only one specific high affinity ligand, F2L, and a more restricted tissue/cell distribution. In this study, we showed that FPR2/ALX behaved as the prototypical receptor FPR1. The agonist-dependent phosphorylation used a hierarchical mechanism with a prominent role of Ser(329), Thr(332), and Thr(335). Phosphorylation of FPR2/ALX was essential for its desensitization but the lack of phosphorylation did not result in enhanced or sustained responses. In contrast, resting FPR3 displayed a marked level of phosphorylation, which was only slightly increased upon agonist stimulation. Another noticeable difference between the two receptors was their subcellular distribution in unstimulated cells. Although FPR2/ALX was evenly distributed at the plasma membrane FPR3 was localized in small intracellular vesicles. By swapping domains between FPR2/ALX and FPR3, we uncovered the determinants involved in the basal phosphorylation of FPR3. Experiments aimed at monitoring receptor-bound antibody uptake showed that the intracellular distribution of FPR3 resulted from a constitutive internalization that was independent of C terminus phosphorylation. Unexpectedly, exchanging residues 1 to 53, which encompass the N-terminal extracellular region and the first transmembrane domain, between FPR2/ALX and FPR3 switched localization of the receptors from the plasma membrane to intracellular vesicles and vice versa. A clathrin-independent, possibly caveolae-dependent, mechanism was involved in FPR3 constitutive internalization. The peculiar behavior of FPR3 most probably serves distinct physiological functions that remain largely unknown.


Subject(s)
Caveolae/metabolism , Gene Expression Regulation/physiology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , HEK293 Cells , HL-60 Cells , Humans , Mutation, Missense , Phosphorylation/physiology , Protein Structure, Tertiary , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics
6.
Cell Host Microbe ; 7(6): 463-73, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20542250

ABSTRACT

Virulence of emerging community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) and other highly pathogenic S. aureus strains depends on their production of phenol-soluble modulin (PSM) peptide toxins, which combine the capacities to attract and lyse neutrophils. The molecular basis of PSM-stimulated neutrophil recruitment has remained unclear. Here, we demonstrate that the human formyl peptide receptor 2 (FPR2/ALX), which has previously been implicated in control of endogenous inflammatory processes, senses PSMs at nanomolar concentrations and initiates proinflammatory neutrophil responses to CA-MRSA. Specific blocking of FPR2/ALX or deletion of PSM genes in CA-MRSA severely diminished neutrophil detection of CA-MRSA. Furthermore, a specific inhibitor of FPR2/ALX and of its functional mouse counterpart blocked PSM-mediated leukocyte infiltration in vivo in a mouse model. Thus, the innate immune system uses a distinct FPR2/ALX-dependent mechanism to specifically sense bacterial peptide toxins and detect highly virulent bacterial pathogens. FPR2/ALX represents an attractive target for new anti-infective or anti-inflammatory strategies.


Subject(s)
Bacterial Toxins/metabolism , Host-Pathogen Interactions , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Neutrophils/immunology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Animals , Community-Acquired Infections/microbiology , Female , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Mice , Mice, Inbred BALB C , Models, Biological , Neutrophils/microbiology , Protein Binding , Receptors, Formyl Peptide/physiology , Staphylococcal Infections
7.
J Biol Chem ; 285(19): 14338-45, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20220135

ABSTRACT

The N-terminal part of the calcium-regulated and phospholipid-binding protein annexin AI contains peptide sequences with pro- and anti-inflammatory activities. We have earlier shown that a proinflammatory signal triggered by one of these peptides, Gln(9)-Lys(25), is mediated by FPR1, a member of the formyl peptide receptor family expressed in human neutrophils. To determine the core structure in Gln(9)-Lys(25), smaller peptides were generated, and their capacity to activate neutrophils was determined. A peptide spanning from amino acid Glu(14) to Lys(25) was inactive, whereas the activity was retained in the Gln(9)-Tyr(20) peptide. Removal of amino acids from the C and N terminus of Gln(9)-Tyr(20) revealed that the first amino acid (Gln(9)) was of the utmost importance for activity. The core structure that activated the neutrophil NADPH oxidase to release superoxide anions was Gln(9)-Ala(10)-Trp(11)-Phe(12). This peptide also inhibited the activity induced by N-formyl-Met-Leu-Phe and WKYMVM. A structural model of the peptide agonist-FPR1 complex suggests that the transmembrane part of the binding pocket of the receptor binds optimally to a tetrapeptide. According to the model and the results presented, the N-terminal amino acid glutamine in Gln(9)-Phe(12) is located close to the bottom of the binding cleft, leaving for steric reasons insufficient space to extend the peptide at the N terminus. The addition of amino acids at the C terminus will not affect binding. The model presented may be helpful in developing specific FPR1 ligands.


Subject(s)
Annexin A1/chemistry , Annexin A1/metabolism , Neutrophils/enzymology , Oligopeptides/chemistry , Oligopeptides/metabolism , Receptors, Formyl Peptide/metabolism , Acetylation , Adult , Calcium/metabolism , Computational Biology , Cytosol/metabolism , HL-60 Cells , Humans , Models, Molecular , NADPH Oxidases/metabolism , Protein Conformation , Superoxides/metabolism
8.
Microbes Infect ; 12(5): 415-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20156579

ABSTRACT

The biosynthesis of proteins with N-terminal formylated methionine residues and subsequent protein deformylation are unique and invariant bacterial processes. They are exploited by the capacity of the human innate immune system to sense formylated peptides (FPs) and targeted by the deformylation-blocking antibiotic actinonin. We show that human polymorphonuclear leukocytes respond via the formyl peptide receptor (FPR) with increased calcium ion fluxes, chemotactic migration, IL-8 release, and CD11b upregulation to the human pathogen Staphylococcus aureus upon actinonin treatment. These data underscore the crucial role of bacterial FPs in innate immunity and indicate that deformylase inhibition may have considerable proinflammatory consequences.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Inflammation , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/physiology , Staphylococcus aureus/enzymology , Staphylococcus aureus/immunology , CD11b Antigen/biosynthesis , Calcium/metabolism , Chemotaxis , Enzyme Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Interleukin-8/metabolism , Neutrophils/immunology , Staphylococcus aureus/drug effects
9.
Mol Pharmacol ; 77(2): 159-70, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19903830

ABSTRACT

N-formyl peptide receptor (FPR1) and N-formyl peptide receptor-like 1 (FPRL1, now known as FPR2) are G protein-coupled receptors involved in host defense and sensing cellular dysfunction. Because of the potential for FPR1/FPR2 as a therapeutic target, our recent high-throughput screening efforts have focused on the identification of unique nonpeptide agonists of FPR1/FPR2. In the present studies, we screened a chemolibrary of drug-like molecules for their ability to induce intracellular calcium mobilization in RBL-2H3 cells transfected with human FPR1 or FPR2. Screening of these compounds resulted in the identification of novel and potent agonists that activated both FPR1 and FPR2, as well as compounds that were specific for either FPR1 or FPR2 with EC(50) values in the low micromolar range. Specificity of the compounds was supported by analysis of calcium mobilization in HL-60 cells transfected with human FPR1 and FPR2. In addition, all but one agonist activated intracellular calcium flux and chemotaxis in human neutrophils, irrespective of agonist specificity for FPR1 or FPR2. Molecular modeling of the group of FPR1 and FPR2 agonists using field point methodology allowed us to create pharmacophore models for ligand binding sites and formulate requirements for these specific N-formyl peptide receptor agonists. These studies further demonstrate that agonists of FPR1/FPR2 include compounds with wide chemical diversity and that analysis of such compounds can enhance our understanding of their ligand/receptor interaction.


Subject(s)
Models, Molecular , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/metabolism , Small Molecule Libraries/metabolism , Animals , Binding Sites/physiology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Protein Structure, Secondary , Rats , Receptors, Formyl Peptide/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
10.
J Med Chem ; 52(16): 5044-57, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19639995

ABSTRACT

Following a ligand-based drug design approach, a potent mixed formyl peptide receptor 1 (FPR1) and formyl peptide receptor-like 1 (FPRL1) agonist (14a) and a potent and specific FPRL1 agonist (14x) were identified. These compounds belong to a large series of pyridazin-3(2H)-one derivatives substituted with a methyl group at position 6 and a methoxy benzyl at position 4. At position 2, an acetamide side chain is essential for activity. Likewise, the presence of lipophilic and/or electronegative substituents in the position para to the aryl group at the end of the chain plays a critical role for activity. Affinity for FPR1 receptors was evaluated by measuring intracellular calcium flux in HL-60 cells transfected with FPR1, FPRL1, and FPRL2. Agonists were able to activate intracellular calcium mobilization and chemotaxis in human neutrophils. The most potent chemotactic agent (EC(50) = 0.6 microM) was the mixed FPR/FPRL1 agonist 14h.


Subject(s)
Pyridazines/chemical synthesis , Receptors, Formyl Peptide/agonists , Receptors, Lipoxin/agonists , Calcium/metabolism , Chemotaxis, Leukocyte/drug effects , HL-60 Cells , Humans , In Vitro Techniques , Neutrophils/drug effects , Neutrophils/physiology , Pyridazines/chemistry , Pyridazines/pharmacology , Structure-Activity Relationship
11.
Cell Calcium ; 45(5): 431-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19282028

ABSTRACT

Human neutrophils express formyl peptide receptor 1 and 2 (FPR1 and FPR2), two highly homologous G-protein-coupled cell surface receptors important for the cellular recognition of chemotactic peptides. They share many functional as well as signal transduction features, but some fundamental differences have been described. One such difference was recently presented when the FPR2-specific ligand MMK-1 was shown to trigger a unique signal in neutrophils [S. Partida-Sanchez, P. Iribarren, M.E. Moreno-Garcia, et al., Chemotaxis and calcium responses of phagocytes to formyl peptide receptor ligands is differentially regulated by cyclic ADP ribose, J. Immunol. 172 (2004) 1896-1906]. This signal bypassed the emptying of the intracellular calcium stores, a route normally used to open the store-operated calcium channels present in the plasma membrane of neutrophils. Instead, the binding of MMK-1 to FPR2 was shown to trigger a direct opening of the plasma membrane channels. In this report, we add MMK-1 to a large number of FPR2 ligands that activate the neutrophil superoxide-generating NADPH-oxidase. In contrast to earlier findings we show that the transient rise in intracellular free calcium induced by MMK-1 involves both a release of calcium from intracellular stores and an opening of channels in the plasma membrane. The same pattern was obtained with another characterized FPR2 ligand, WKYMVM, and it is also obvious that the two formyl peptide receptor family members trigger the same type of calcium response in human neutrophils.


Subject(s)
Calcium Channels/metabolism , Cell Membrane/metabolism , NADPH Oxidases/metabolism , Neutrophils/metabolism , Peptides/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Signal Transduction/physiology , Calcium/metabolism , Enzyme Activation , HL-60 Cells , Humans , N-Formylmethionine Leucyl-Phenylalanine/metabolism , NADPH Oxidases/genetics , Neutrophils/cytology , Peptides/genetics , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Superoxides/metabolism
12.
J Biol Chem ; 283(45): 31038-46, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18772131

ABSTRACT

Most G-protein-coupled receptors (GPCRs) form di(oligo)-meric structures that constitute signaling and trafficking units and might be essential for receptor functions. Cell responses to complement C5a receptor (C5aR) are tightly controlled by receptor desensitization and internalization. To examine the implication of dimerization in C5aR regulation, we generated an NH(2)-terminally modified C5aR mutant, unable to bind C5a, and a phosphorylation-deficient mutant. Neither an intact NH(2) terminus nor the presence of COOH-terminal phosphorylation sites appeared to be required for the formation of C5aR dimers. Upon C5a stimulation, mutant receptors did not internalize when individually expressed. C5a stimulation of cells that co-expressed wild type C5aR together with either unliganded or phosphorylation-deficient mutant resulted in co-internalization of mutant receptors with C5aR. Unliganded GPCRs can be cross-phosphorylated within a heterologous receptor dimer or by second messenger-activated kinases. C5a stimulation of (32)P-labeled cells that co-expressed the unliganded mutant with either C5aR or the phosphorylation-deficient mutant did not induce phosphorylation of the unliganded mutant. We can thus postulate that, in the case of C5aR, the stimulation and phosphorylation of one monomer is enough to lead to dimer internalization. The existence and functional implication of di(oligo)mer formation may be important for an accurate C5aR down-regulation in pathological conditions.


Subject(s)
Down-Regulation/physiology , Receptor Aggregation/physiology , Receptors, Complement/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Line , Complement C5a/genetics , Complement C5a/metabolism , Dimerization , Humans , Mutation , Phosphorylation/genetics , Receptor, Anaphylatoxin C5a , Receptors, Complement/genetics , Receptors, G-Protein-Coupled/genetics
13.
J Leukoc Biol ; 83(2): 245-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17984291

ABSTRACT

Serum amyloid A (SAA) is one of the acute-phase reactants, a group of plasma proteins that increases immensely in concentration during microbial infections and inflammatory conditions, and a close relationship between SAA levels and disease activity in rheumatoid arthritis (RA) has been observed. RA is an inflammatory disease, where neutrophils play important roles, and SAA is thought to participate in the inflammatory reaction by being a neutrophil chemoattractant and inducer of proinflammatory cytokines. The biological effects of SAA are reportedly mediated mainly through formyl peptide receptor like-1 (FPRL1), a G protein-coupled receptor (GPCR) belonging to the formyl peptide receptor family. Here, we confirmed the affinity of SAA for FPRL1 by showing that stably transfected HL-60 cells expressing FPRL1 were activated by SAA and that the response was inhibited by the use of the FPRL1-specific antagonist WRWWWW (WRW4). We also show that SAA activates the neutrophil NADPH-oxidase and that a reserve pool of receptors is present in storage organelles mobilized by priming agents such as TNF-alpha and LPS from Gram-negative bacteria. The induced activity was inhibited by pertussis toxin, indicating the involvement of a GPCR. However, based on FPRL1-specific desensitization and use of FPRL1 antagonist WRW4, we found the SAA-mediated effects in neutrophils to be independent of FPRL1. Based on these findings, we conclude that SAA signaling in neutrophils is mediated through a GPCR, distinct from FPRL1. Future identification and characterization of the SAA receptor could lead to development of novel, therapeutic targets for treatment of RA.


Subject(s)
Neutrophils/drug effects , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Lipoxin/physiology , Serum Amyloid A Protein/physiology , Signal Transduction/physiology , Calcium Signaling/drug effects , Enzyme Activation/drug effects , Gelsolin/pharmacology , HL-60 Cells , Humans , Lipopolysaccharides/pharmacology , NADPH Oxidases/metabolism , Neutrophils/enzymology , Neutrophils/metabolism , Oligopeptides/pharmacology , Organelles/metabolism , Peptide Fragments/pharmacology , Pertussis Toxin/pharmacology , Phosphatidylinositol 4,5-Diphosphate/physiology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/genetics , Receptors, G-Protein-Coupled/drug effects , Receptors, Lipoxin/antagonists & inhibitors , Receptors, Lipoxin/genetics , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/physiology , Respiratory Burst/drug effects , Serum Amyloid A Protein/pharmacology , Signal Transduction/drug effects , Transfection , Tumor Necrosis Factor-alpha/pharmacology
14.
Cell Signal ; 19(9): 1939-48, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17594911

ABSTRACT

The N-formyl peptide receptor-like 1 (FPRL1) is a G protein-coupled receptor (GPCR) that transmits intracellular signals in response to a variety of agonists, many of them being clearly implicated in human pathology. beta-arrestins are adaptor proteins that uncouple GPCRs from G protein and regulate receptor internalization. They can also function as signal transducers through the scaffolding of signaling molecules, such as components of the extracellular signal-regulated kinase (ERK) cascade. We investigated the role of beta-arrestins in ligand-induced FPRL1 internalization and signaling. In HEK293 cells expressing FPRL1, fluorescence microscopy revealed that agonist-stimulated FPRL1 remained co-localized with beta-arrestins during endocytosis. Internalization of FPRL1, expressed in a mouse embryonic fibroblast (MEF) cell line lacking endogenous beta-arrestins, was highly compromised. This distinguishes FPRL1 from the prototypical formyl peptide receptor FPR that is efficiently internalized in the absence of beta-arrestins. In both HEK293 and MEF cells, FPRL1-mediated ERK1/2 activation was a rapid and transient event. The kinetics and extent of ERK1/2 activation were not significantly modified by beta-arrestin overexpression. The pattern of FPRL1-mediated ERK1/2 activation was similar whether cells express or not beta-arrestins. Furthermore, treatment of the FPRL1 expressing cells with pertussis toxin inhibited ERK1/2 activation in MEF and in HEK293 cells. These results led us to conclude that activation of ERK1/2 mediated by FPRL1 occurs primarily through G protein signaling. Since beta-arrestin-mediated signaling has been observed essentially for receptors coupled to G proteins other than G(i), this may be a characteristic of G(i) protein-coupled chemoattractant receptors.


Subject(s)
Arrestins/metabolism , Endocytosis , Receptors, Formyl Peptide/metabolism , Signal Transduction , Animals , Arrestins/deficiency , Cell Line , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/enzymology , GTP-Binding Proteins/metabolism , Gene Expression , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Transport , Recombinant Fusion Proteins/metabolism , beta-Arrestins
15.
Eur J Immunol ; 37(7): 1966-77, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17559171

ABSTRACT

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.


Subject(s)
Antigens, Dermatophagoides/immunology , Eosinophils/immunology , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/immunology , Signal Transduction/immunology , Animals , Betula/immunology , Calcium/metabolism , Chemotaxis, Leukocyte/immunology , Eosinophils/metabolism , HL-60 Cells , Humans , Pollen/immunology , Polymerase Chain Reaction , RNA, Messenger/analysis , Receptors, CCR3 , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Leukotriene B4/immunology , Receptors, Leukotriene B4/metabolism , Receptors, Lipoxin/metabolism , Transfection
16.
Biochimie ; 89(9): 1089-106, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17428601

ABSTRACT

Leukocyte recruitment to sites of inflammation and infection is dependent on the presence of a gradient of locally produced chemotactic factors. This review is focused on current knowledge about the activation and regulation of chemoattractant receptors. Emphasis is placed on the members of the N-formyl peptide receptor family, namely FPR (N-formyl peptide receptor), FPRL1 (FPR like-1) and FPRL2 (FPR like-2), and the complement fragment C5a receptors (C5aR and C5L2). Upon chemoattractant binding, the receptors transduce an activation signal through a G protein-dependent pathway, leading to biochemical responses that contribute to physiological defense against bacterial infection and tissue damage. C5aR, and the members of the FPR family that were previously thought to be restricted to phagocytes proved to have a much broader spectrum of cell expression. In addition to N-formylated peptides, numerous unrelated ligands were recently found to interact with FPR and FPRL1. Novel agonists include both pathogen- and host-derived components, and synthetic peptides. Antagonistic molecules have been identified that exhibit limited receptor specificity. How distinct ligands can both induce different biological responses and produce different modes of receptor activation and unique sets of cellular responses are discussed. Cell responses to chemoattractants are tightly regulated at the level of the receptors. This review describes in detail the regulation of receptor signalling and the multi-step process of receptor inactivation. New concepts, such as receptor oligomerization and receptor clustering, are considered. Although FPR, FPRL1 and C5aR trigger similar biological functions and undergo a rapid chemoattractant-mediated phosphorylation, they appear to be differentially regulated and experience different intracellular fates.


Subject(s)
Receptor, Anaphylatoxin C5a/physiology , Receptors, Formyl Peptide/physiology , Amino Acid Sequence , Animals , Humans , Models, Biological , Molecular Sequence Data , Phosphorylation , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Sequence Homology, Amino Acid , Signal Transduction
17.
Eur J Immunol ; 35(8): 2486-95, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16025565

ABSTRACT

N-formyl peptides are cleavage products of bacterial and mitochondrial proteins, and can attract leukocytes to sites of infection or tissue damage. In this study, HL-60 cell lines expressing the human N-formyl peptide receptor FPR or its two homologues (FPRL1, FPRL2) were used to determine the receptor selectivity of N-formylated peptides derived from Listeria monocytogenes or from human mitochondrial proteins. Bacterial peptides were 100-fold more potent on FPR than on FPRL1, whereas none of them could trigger intracellular signaling through FPRL2. In contrast, N-formylated hexapeptides corresponding to the N terminus of mitochondrial NADH dehydrogenase subunits 4 (fMLKLIV) and 6 (fMMYALF), and cytochrome c oxidase subunit I (fMFADRW) were equally potent on FPR and FPRL1. They triggered cellular responses with the following order of potency: fMMYALF > fMLKLIV > fMFADRW, with an EC50, in a Fura-2 calcium mobilization assay, of 10 nM, 44 nM, and 160 nM on FPR-expressing cells, and 15 nM, 55 nM and 120 nM on FPRL1-expressing cells. fMMYALF was also a low-affinity agonist of FPRL2 (EC50 of 1 microM) and was chemotactic for both FPRL1- and FPRL2-expressing cells. We identified novel mitochondrial host-derived agonists for human N-formyl-peptide receptors that might play a role in inflammatory or degenerative processes linked to their stimulation.


Subject(s)
Listeria monocytogenes/metabolism , Mitochondria/metabolism , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/agonists , Receptors, Lipoxin/metabolism , Calcium/metabolism , Chemotaxis/physiology , HL-60 Cells , Humans , Mitogen-Activated Protein Kinases/metabolism , NADH Dehydrogenase/physiology , Peptide Fragments/physiology , Superoxides/metabolism
19.
Biochem J ; 372(Pt 2): 453-63, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12600272

ABSTRACT

Migration of myeloid cells towards a source of chemoattractant, such as the C5a anaphylatoxin, is triggered by the activation of a G-protein-coupled receptor. In the present study, we have used a yeast two-hybrid approach to find unknown partners of the C5a receptor (C5aR). Using the cytosolic C-terminal region of C5aR as bait to screen a human leucocyte cDNA library, we identified the Wiskott-Aldrich syndrome protein (WASP) as a potential partner of C5aR. WASP is known to have an essential function in regulating actin dynamics at the cell leading edge. The interaction was detected with both the fragment of WASP containing amino acids 1-321 (WASP.321) and WASP with its actin-nucleation-promoting domain [verprolin-like, central and acidic (VCA) domain] deleted. The interaction between C5aR and the WASP.321 was supported further by an in vitro binding assay between a radiolabelled WASP.321 fragment and a receptor C-terminus glutathione S-transferase (GST) fusion protein, as well as by GST pull-down, co-immunoprecipitation and immunofluorescence experiments. In the yeast two-hybrid assay, full-length WASP showed no ability to interact with the C-terminal domain of C5aR. This is most probably due to an auto-inhibited conformation imposed by the VCA domain. In HEK-293T cells co-transfected with full-length WASP and C5aR, only a small amount of WASP was co-precipitated with the receptor. However, in the presence of the active form of the GTPase Cdc42 (Cdc42V12), which is thought to switch WASP to an active 'open conformation', the amount of WASP associated with the receptor was markedly increased. We hypothesize that a transient interaction between C5aR and WASP occurs following the stimulation of C5aR and Cdc42 activation. This might be one mechanism by which WASP is targeted to the plasma membrane and by which actin assembly is spatially controlled in cells moving in a gradient of C5a.


Subject(s)
Antigens, CD/metabolism , Peptide Fragments/metabolism , Proteins/metabolism , Receptors, Complement/metabolism , Actins/chemistry , Actins/metabolism , Animals , Antigens, CD/genetics , Binding Sites , Blotting, Western , COS Cells/cytology , Cell Membrane , Cells, Cultured , Chlorocebus aethiops , Complement C5a/metabolism , Enzyme Activation , Fluorescent Antibody Technique , Gene Deletion , Gene Library , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Leukocytes/metabolism , Mutation , Peptide Fragments/genetics , Precipitin Tests , Protein Binding , Protein Conformation , Proteins/genetics , Receptor, Anaphylatoxin C5a , Receptors, Complement/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transfection , Two-Hybrid System Techniques , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome Protein , cdc42 GTP-Binding Protein/metabolism
20.
Blood ; 100(5): 1835-44, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12176907

ABSTRACT

A tetracycline-controlled expression system was adapted to the human promyelocytic HL-60 cell line by placement of the transactivator (tTA-off) sequence under the control of the human EF-1alpha promoter region. Constitutively active and dominant-inhibitory forms of Cdc42 (Cdc42V12 and Cdc42N17, respectively) were conditionally expressed in this system. The expression of Cdc42V12 had no marked effect on chemoattractant-mediated superoxide production, corroborating previous results indicating that the guanosine 5'-triphosphate (GTP)-bound form of Cdc42 is ineffective in directly activating nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in a cell-free system. However, the N17 mutant potently inhibited chemoattractant-induced superoxide production. The expression of Cdc42N17 interfered with the GTP-loading of Rac and Ras and with the activation of the MAP-kinase pathway. A drastic reduction of chemoattractant-induced inositol-1,4,5-trisphosphate formation and calcium mobilization was observed, corroborating previous in vitro study results identifying PLCbeta2 as a Rac/Cdc42 effector. Cdc42N17 was also found to inhibit the translocation of Ras-GRF2, a guanine nucleotide exchange factor for Ras and Rac but not for Cdc42. Thus, the dominant-inhibitory mutant Cdc42N17 was found to interfere at multiple levels in the signaling pathways. The pleiotropic inhibitory effects of Cdc42N17 illustrate the potential pitfalls of using dominant-inhibitory proteins to study the function of Ras-family GTPases. In this regard, a number of conclusions drawn from the use of dominant-inhibitory mutants in myeloid cells might have to be reconsidered.


Subject(s)
NADPH Oxidases/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , Enzyme Activation , HL-60 Cells , Humans , Promoter Regions, Genetic , Tetracycline/metabolism , Transcriptional Activation , cdc42 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...