253 results on '"Ye, RD"'
Search Results
102. Structural determinants for the interaction of formyl peptide receptor 2 with peptide ligands.
- Author
-
He HQ, Troksa EL, Caltabiano G, Pardo L, and Ye RD
- Subjects
- Amino Acid Substitution, Humans, Ligands, Mutagenesis, Site-Directed, Mutation, Missense, N-Formylmethionine Leucyl-Phenylalanine chemistry, N-Formylmethionine Leucyl-Phenylalanine metabolism, Receptors, CXCR4 chemistry, Receptors, CXCR4 genetics, Receptors, CXCR4 metabolism, Receptors, Formyl Peptide genetics, Receptors, Formyl Peptide metabolism, Receptors, Lipoxin genetics, Receptors, Lipoxin metabolism, Structural Homology, Protein, Structure-Activity Relationship, Molecular Dynamics Simulation, N-Formylmethionine Leucyl-Phenylalanine analogs & derivatives, Receptors, Formyl Peptide chemistry, Receptors, Lipoxin chemistry
- Abstract
Unlike formyl peptide receptor 1 (FPR1), FPR2/ALX (FPR2) interacts with peptides of diverse sequences but has low affinity for the Escherichia coli-derived chemotactic peptide fMet-Leu-Phe (fMLF). Using computer modeling and site-directed mutagenesis, we investigated the structural requirements for FPR2 to interact with formyl peptides of different length and composition. In calcium flux assay, the N-formyl group of these peptides is necessary for activation of both FPR2 and FPR1, whereas the composition of the C-terminal amino acids appears more important for FPR2 than FPR1. FPR2 interacts better with pentapeptides (fMLFII, fMLFIK) than tetrapeptides (fMLFK, fMLFW) and tripeptide (fMLF) but only weakly with peptides carrying negative charges at the C terminus (e.g. fMLFE). In contrast, FPR1 is less sensitive to negative charges at the C terminus. A CXCR4-based homology model of FPR1 and FPR2 suggested that Asp-281(7.32) is crucial for the interaction of FPR2 with certain formyl peptides as its negative charge may be repulsive with the terminal COO- group of fMLF and negatively charged Glu in fMLFE. Asp-281(7.32) might also form a stable interaction with the positively charged Lys in fMLFK. Site-directed mutagenesis was performed to remove the negative charge at position 281 in FPR2. The D281(7.32)G mutant showed improved affinity for fMLFE and fMLF and reduced affinity for fMLFK compared with wild type FPR2. These results indicate that different structural determinants are used by FPR1 and FPR2 to interact with formyl peptides.
- Published
- 2014
- Full Text
- View/download PDF
103. Detection of intact transcription factors in human neutrophils.
- Author
-
McDonald PP and Ye RD
- Subjects
- Cell Nucleus genetics, Cell Nucleus metabolism, DNA metabolism, Electrophoretic Mobility Shift Assay, Humans, Protein Binding, Neutrophils metabolism, Transcription Factors metabolism
- Abstract
The crucial contribution of neutrophils to innate immunity extends well beyond their traditional role as professional phagocytes. Indeed, it is now well established that neutrophils generate a plethora of inflammatory cytokines and chemokines that are profoundly involved in the onset and evolution of the inflammatory reaction. Several recent studies have also shown that neutrophils can represent an important source of inflammatory cytokines in a number of pathophysiological settings. The inflammatory cytokines produced by neutrophils are generally encoded by immediate-early response genes, which in turn depend on the activation of transcription factors such as those belonging to the nuclear factor-kappa B and signal transducers and activators of transcription families. We have shown in the past that such factors are expressed and inducible in neutrophils stimulated by physiological agonists. However, the detection of intact (i.e., undegraded) transcription factors in neutrophils requires special precautions and an alternative protocol due to the huge amounts of endogenous proteases present in these cells. This protocol is the focus of this chapter.
- Published
- 2014
- Full Text
- View/download PDF
104. Serum amyloid a differentially activates microglia and astrocytes via the PI3K pathway.
- Author
-
Yu Y, Liu J, Li SQ, Peng L, and Ye RD
- Subjects
- Animals, Apoptosis drug effects, CD11b Antigen metabolism, Cell Cycle drug effects, Cells, Cultured, Cytokines genetics, Cytokines metabolism, Dose-Response Relationship, Drug, Glial Fibrillary Acidic Protein metabolism, Humans, Mice, Mice, Inbred C57BL, Microglia enzymology, Statistics, Nonparametric, Time Factors, Astrocytes drug effects, Astrocytes enzymology, Microglia drug effects, Phosphatidylinositol 3-Kinases metabolism, Serum Amyloid A Protein pharmacology, Signal Transduction drug effects
- Abstract
Microglia and astrocytes in the brain play an important role in the development and progression of Alzheimer's disease (AD). Serum amyloid A (SAA) is a major acute-phase protein produced locally in the brain and colocalizes with senile plaques in AD patients. We investigated whether SAA plays a role in the development of AD. The viability of cultured primary microglia and astrocytes was measured by MTT; cell cycle and apoptosis analysis was also conducted. Cultured microglia and astrocytes were stimulated with 1 μM SAA for different periods of time (2, 4, 6, 12 h) or treated with 1 μM SAA with or without 15 min pretreatment of MAPK or PI3K inhibitors. Total RNA was extracted for qPCR analysis. SAA induced morphological changes of primary microglia but not astrocytes. Interestingly, SAA increased the viability of microglia by inhibiting their apoptosis and reduced the viability of astrocytes by inducing G1 cell cycle arresting. SAA treatment increased the mRNA levels of IL-6, TNF-α, IL12p40, IL23p19, and IL-10, with higher potency in microglia than in astrocytes. However, SAA induced more iNOS mRNA in astrocytes than in microglia. SAA induced these cytokines and iNOS expression by activating the PI3K pathway in both glial cells, but selectively activated the JNK pathway in microglia and the NF-κB pathway in astrocytes. These results suggest that SAA can stimulate a different reactive phenotype in microglia and astrocytes, and SAA regulates cell viability differently in these two glial cells in part through the PI3K pathway.
- Published
- 2014
- Full Text
- View/download PDF
105. The Concise Guide to PHARMACOLOGY 2013/14: overview.
- Author
-
Alexander SP, Benson HE, Faccenda E, Pawson AJ, Sharman JL, McGrath JC, Catterall WA, Spedding M, Peters JA, Harmar AJ, Abul-Hasn N, Anderson CM, Anderson CM, Araiksinen MS, Arita M, Arthofer E, Barker EL, Barratt C, Barnes NM, Bathgate R, Beart PM, Belelli D, Bennett AJ, Birdsall NJ, Boison D, Bonner TI, Brailsford L, Bröer S, Brown P, Calo G, Carter WG, Catterall WA, Chan SL, Chao MV, Chiang N, Christopoulos A, Chun JJ, Cidlowski J, Clapham DE, Cockcroft S, Connor MA, Cox HM, Cuthbert A, Dautzenberg FM, Davenport AP, Dawson PA, Dent G, Dijksterhuis JP, Dollery CT, Dolphin AC, Donowitz M, Dubocovich ML, Eiden L, Eidne K, Evans BA, Fabbro D, Fahlke C, Farndale R, Fitzgerald GA, Fong TM, Fowler CJ, Fry JR, Funk CD, Futerman AH, Ganapathy V, Gaisnier B, Gershengorn MA, Goldin A, Goldman ID, Gundlach AL, Hagenbuch B, Hales TG, Hammond JR, Hamon M, Hancox JC, Hauger RL, Hay DL, Hobbs AJ, Hollenberg MD, Holliday ND, Hoyer D, Hynes NA, Inui KI, Ishii S, Jacobson KA, Jarvis GE, Jarvis MF, Jensen R, Jones CE, Jones RL, Kaibuchi K, Kanai Y, Kennedy C, Kerr ID, Khan AA, Klienz MJ, Kukkonen JP, Lapoint JY, Leurs R, Lingueglia E, Lippiat J, Lolait SJ, Lummis SC, Lynch JW, MacEwan D, Maguire JJ, Marshall IL, May JM, McArdle CA, McGrath JC, Michel MC, Millar NS, Miller LJ, Mitolo V, Monk PN, Moore PK, Moorhouse AJ, Mouillac B, Murphy PM, Neubig RR, Neumaier J, Niesler B, Obaidat A, Offermanns S, Ohlstein E, Panaro MA, Parsons S, Pwrtwee RG, Petersen J, Pin JP, Poyner DR, Prigent S, Prossnitz ER, Pyne NJ, Pyne S, Quigley JG, Ramachandran R, Richelson EL, Roberts RE, Roskoski R, Ross RA, Roth M, Rudnick G, Ryan RM, Said SI, Schild L, Sanger GJ, Scholich K, Schousboe A, Schulte G, Schulz S, Serhan CN, Sexton PM, Sibley DR, Siegel JM, Singh G, Sitsapesan R, Smart TG, Smith DM, Soga T, Stahl A, Stewart G, Stoddart LA, Summers RJ, Thorens B, Thwaites DT, Toll L, Traynor JR, Usdin TB, Vandenberg RJ, Villalon C, Vore M, Waldman SA, Ward DT, Willars GB, Wonnacott SJ, Wright E, Ye RD, Yonezawa A, and Zimmermann M
- Subjects
- Humans, Ligands, Pharmaceutical Preparations chemistry, Databases, Pharmaceutical, Molecular Targeted Therapy, Pharmacology
- Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties from the IUPHAR database. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. This compilation of the major pharmacological targets is divided into seven areas of focus: G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors & Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and GRAC and provides a permanent, citable, point-in-time record that will survive database updates., (Copyright © 2013 The British Pharmacological Society.)
- Published
- 2013
- Full Text
- View/download PDF
106. Pharmacology in China: a brief overview.
- Author
-
Wang MW, Ye RD, and Zhu Y
- Subjects
- China, Clinical Trials as Topic trends, Drug Industry, History, 20th Century, History, 21st Century, Pharmacology trends, Societies, Scientific, Pharmacology history, Pharmacology methods
- Published
- 2013
- Full Text
- View/download PDF
107. Bioluminescent detection of peroxynitrite with a boronic acid-caged luciferin.
- Author
-
Sieracki NA, Gantner BN, Mao M, Horner JH, Ye RD, Malik AB, Newcomb ME, and Bonini MG
- Subjects
- Animals, Boronic Acids, Mice, Nitric Oxide Synthase Type II physiology, Peroxynitrous Acid blood, Benzothiazoles metabolism, Luminescent Measurements methods, Peroxynitrous Acid analysis
- Abstract
Peroxynitrite, a highly reactive biological oxidant, is formed under pathophysiologic conditions from the diffusion-limited reaction of nitric oxide and superoxide radical anion. Peroxynitrite has been implicated as the mediator of nitric oxide toxicity in many diseases and as an important signaling disrupting molecule (L. Liaudet et al., Front. Biosci.14, 4809-4814, 2009) [1]. Biosensors effective at capturing peroxynitrite in a specific and fast enough manner for detection, along with readouts compatible with in vivo studies, are lacking. Here we report that the boronic acid-based bioluminescent system PCL-1 (peroxy-caged luciferin-1), previously reported as a chemoselective sensor for hydrogen peroxide (G.C. Van de Bittner et al., Proc. Natl. Acad. Sci. USA107, 21316-21321, 2010) [2], reacts with peroxynitrite stoichiometrically with a rate constant of 9.8±0.3×10(5)M(-1)s(-1) and a bioluminescence detection limit of 16nM, compared to values of 1.2±0.3M(-1)s(-1) and 231nM for hydrogen peroxide. Further, we demonstrate bioluminescent detection of peroxynitrite in the presence of physiological competitors: carbon dioxide, glutathione, albumin, and catalase. We also demonstrate the utility of this method to assess peroxynitrite formation in mammalian cells by measuring peroxynitrite generated under normal culture conditions after stimulation of macrophages with bacterial endotoxin lipopolysaccharide. Thus, the PCL-1 method for measuring peroxynitrite generation shows superior selectivity over other oxidants under in vivo conditions., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
108. V101L of human formyl peptide receptor 1 (FPR1) increases receptor affinity and augments the antagonism mediated by cyclosporins.
- Author
-
Zhou C, Zhou Y, Wang J, Feng Y, Wang H, Xue J, Chen Y, Ye RD, and Wang MW
- Subjects
- Amino Acid Substitution, Animals, Asian People, CHO Cells drug effects, Calcium metabolism, Chemotaxis drug effects, Cricetinae, Cricetulus, Cyclosporins metabolism, Cyclosporins pharmacology, Female, Haplotypes, Humans, Male, Mitogen-Activated Protein Kinase 3 metabolism, N-Formylmethionine Leucyl-Phenylalanine pharmacology, Pharmacogenetics, Cyclosporine pharmacology, Polymorphism, Single Nucleotide, Receptors, Formyl Peptide genetics, Receptors, Formyl Peptide metabolism
- Abstract
Genetic variation plays a major role in drug response variability. CsA (cyclosporin A), a widely used immunosuppressive agent, is a specific antagonist for FPR1 (formyl peptide receptor 1), which is an important G-protein-coupled chemoattractant receptor in the innate immune system. In order to study the variable responses of cyclosporins to different FPR1 mutants, we investigated the distribution of human FPR1 haplotypes among 209 healthy Han Chinese subjects. The haplotype pattern in Han Chinese were characterized on the basis of five SNPs (single nucleotide polymorphisms), including rs5030878 (p.T11I), rs2070745 (p.V101L), rs5030880 (p.R190W), rs1042229 (p.N192K) and rs867228 (p.A346E). Receptor binding affinity of cyclosporins to FPR1 haplotypes was assessed using N-formyl-Nle-Leu-Phe-Nle-Tyr-Lys-FITC in CHO-G(α16) cells stably transfected with cDNAs encoding the top 12 FPR1 haplotypes in the Han Chinese. Variants of FPR1 carrying a single amino acid substitution of leucine for valine at position 101 (p.Leu(101)) displayed significantly higher pK(i) values for CsA and CsH (cyclosporin H), indicative of an improved receptor affinity. The polymorphism of FPR1 p.Leu(101) also enhanced the inhibitory effects of cyclosporins on fMLF (N-formyl-methionyl-leucyl-phenylalanine)-induced activities, including calcium mobilization, cell chemotaxis and MAPK (mitogen-activated protein kinase) phosphorylation. These results point to a possible complication for clinical use of CsA in patients carrying the p.Leu(101) allele of FPR1.
- Published
- 2013
- Full Text
- View/download PDF
109. The Listeria monocytogenes ChiA chitinase enhances virulence through suppression of host innate immunity.
- Author
-
Chaudhuri S, Gantner BN, Ye RD, Cianciotto NP, and Freitag NE
- Subjects
- Animals, Chitinases genetics, Disease Models, Animal, Down-Regulation, Gene Deletion, Genetic Complementation Test, Listeriosis immunology, Listeriosis microbiology, Mice, Microbial Viability, Nitric Oxide Synthase Type II biosynthesis, Virulence Factors genetics, Chitinases metabolism, Immune Evasion, Immunity, Innate drug effects, Listeria monocytogenes immunology, Listeria monocytogenes pathogenicity, Virulence Factors metabolism
- Abstract
Unlabelled: Environmental pathogens survive and replicate within the outside environment while maintaining the capacity to infect mammalian hosts. For some microorganisms, mammalian infection may be a relatively rare event. Understanding how environmental pathogens retain their ability to cause disease may provide insight into environmental reservoirs of disease and emerging infections. Listeria monocytogenes survives as a saprophyte in soil but is capable of causing serious invasive disease in susceptible individuals. The bacterium secretes virulence factors that promote cell invasion, bacterial replication, and cell-to-cell spread. Recently, an L. monocytogenes chitinase (ChiA) was shown to enhance bacterial infection in mice. Given that mammals do not synthesize chitin, the function of ChiA within infected animals was not clear. Here we have demonstrated that ChiA enhances L. monocytogenes survival in vivo through the suppression of host innate immunity. L. monocytogenes ΔchiA mutants were fully capable of establishing bacterial replication within target organs during the first 48 h of infection. By 72 to 96 h postinfection, however, numbers of ΔchiA bacteria diminished, indicative of an effective immune response to contain infection. The ΔchiA-associated virulence defect could be complemented in trans by wild-type L. monocytogenes, suggesting that secreted ChiA altered a target that resulted in a more permissive host environment for bacterial replication. ChiA secretion resulted in a dramatic decrease in inducible nitric oxide synthase (iNOS) expression, and ΔchiA mutant virulence was restored in NOS2(-/-) mice lacking iNOS. This work is the first to demonstrate modulation of a specific host innate immune response by a bacterial chitinase., Importance: Bacterial chitinases have traditionally been viewed as enzymes that either hydrolyze chitin as a food source or serve as a defense mechanism against organisms containing structural chitin (such as fungi). Recent evidence indicates that bacterial chitinases and chitin-binding proteins contribute to pathogenesis, primarily via bacterial adherence to chitin-like molecules present on the surface of mammalian cells. In contrast, mammalian chitinases have been linked to immunity via inflammatory immune responses that occur outside the context of infection, and since mammals do not produce chitin, the targets of these mammalian chitinases have remained elusive. This work demonstrates that a Listeria monocytogenes-secreted chitinase has distinct functional roles that include chitin hydrolysis and suppression of host innate immunity. The established link between chitinase and the inhibition of host inducible nitric oxide synthase (iNOS) expression may help clarify the thus far elusive relationship observed between mammalian chitinase enzymes and host inflammatory responses occurring in the absence of infection.
- Published
- 2013
- Full Text
- View/download PDF
110. Functional characterization of three mouse formyl peptide receptors.
- Author
-
He HQ, Liao D, Wang ZG, Wang ZL, Zhou HC, Wang MW, and Ye RD
- Subjects
- Animals, Benzamides pharmacology, Calcium metabolism, Cell Line, Tumor, Escherichia coli metabolism, Leukemia, Basophilic, Acute metabolism, Listeria monocytogenes metabolism, Mice, Mitochondria metabolism, Oligopeptides pharmacology, Protein Binding, Quinazolines pharmacology, Rats, Staphylococcus aureus metabolism, Transfection methods, N-Formylmethionine Leucyl-Phenylalanine metabolism, Receptors, Formyl Peptide metabolism
- Abstract
The evolutionary relationship and functional correlation between human formyl peptide receptors (FPRs) and their mouse counterparts remain incompletely understood. We examined three members of the mouse formyl peptide receptor subfamily (mFprs) and found that they differ in agonist preference and cellular distributions. When stably expressed in transfected rat basophilic leukemia (RBL-2H3) cells, mFpr1 was readily activated by N-formylated peptides derived from Listeria monocytogenes (fMIVTLF), Staphylococcus aureus (fMIFL), and mitochondria (fMMYALF). In contrast, the Escherichia coli-derived fMLF was 1000-fold less potent. The aforementioned peptides were much less efficacious at mFpr2, which responded better to the synthetic hexapeptide WKYMVm, the synthetic agonists Quin-C1 (a substituted quinazolinone), and compound 43 (a nitrosylated pyrazolone derivative). Saturation binding assays showed that mFpr1 and mFpr2 were expressed at similar levels on the cell surface, although their affinity for N-formyl-Met-Leu-Phe-Ile-Ile-Lys-fluorescein isothiocyanate varied by more than 1000-fold [dissociation constant (K(d)) values of 2.8 nM for mFpr1 and 4.8 μM for mFpr2]). Contrary to these receptors, mFpr-rs1 responded poorly to all the previously mentioned peptides that were tested. Fluorescent microscopy revealed an intracellular distribution pattern of mFpr-rs1. On the basis of these results, we conclude that mFpr1 is an ortholog of human FPR1 with certain pharmacologic properties of human FPR2/ALX, whereas mFpr2 has much lower affinity for formyl peptides. The intracellular distribution of mFpr-rs1 suggests an evolutionary correlation with human FPR3.
- Published
- 2013
- Full Text
- View/download PDF
111. A critical role for phosphatidylinositol (3,4,5)-trisphosphate-dependent Rac exchanger 1 in endothelial junction disruption and vascular hyperpermeability.
- Author
-
Naikawadi RP, Cheng N, Vogel SM, Qian F, Wu D, Malik AB, and Ye RD
- Subjects
- Animals, Capillary Permeability drug effects, Capillary Permeability genetics, Cell Line, Endothelium, Vascular drug effects, Guanine Nucleotide Exchange Factors deficiency, Guanine Nucleotide Exchange Factors genetics, HL-60 Cells, Humans, Mice, Mice, Knockout, Reactive Oxygen Species metabolism, Signal Transduction genetics, Signal Transduction physiology, Tumor Necrosis Factor-alpha toxicity, Capillary Permeability physiology, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Guanine Nucleotide Exchange Factors physiology
- Abstract
Rationale: The small GTPase Rac is critical to vascular endothelial functions, yet its regulation in endothelial cells remains unclear. Understanding the upstream pathway may delineate Rac activation mechanisms and its role in maintaining vascular endothelial barrier integrity., Objective: By investigating phosphatidylinositol (3,4,5)-trisphosphate-dependent Rac exchanger 1 (P-Rex1), one of the Rac-specific guanine nucleotide exchange factors previously known for G protein-coupled receptor signaling, we sought to determine whether Rac-guanine nucleotide exchange factor is nodal for signal integration and potential target for drug intervention., Methods and Results: Using gene deletion and small interference RNA silencing approach, we investigated the role of P-Rex1 in human lung microvascular endothelial cells. Tumor necrosis factor α (TNF-α) exposure led to disruption of endothelial junctions, and silencing P-Rex1 protected junction integrity. TNF-α stimulated Rac activation and reactive oxygen species production in a P-Rex1-dependent manner. Removal of P-Rex1 significantly reduced intercellular adhesion molecule-1 expression, polymorphonuclear leukocyte transendothelial migration, and leukocyte sequestration in TNF-α-challenged mouse lungs. The P-Rex1 knockout mice were also refractory to lung vascular hyperpermeability and edema in a lipopolysaccharide-induced sepsis model., Conclusions: These results demonstrate for the first time that P-Rex1 expressed in endothelial cells is activated downstream of TNF-α, which is not a G protein-coupled receptor agonist. Our data identify P-Rex1 as a critical mediator of vascular barrier disruption. Targeting P-Rex1 may effectively protect against TNF-α- and lipopolysaccharide-induced endothelial junction disruption and vascular hyperpermeability.
- Published
- 2012
- Full Text
- View/download PDF
112. Activation of Ras-dependent signaling pathways by G(14) -coupled receptors requires the adaptor protein TPR1.
- Author
-
Kwan DH, Yung LY, Ye RD, and Wong YH
- Subjects
- Adaptor Proteins, Vesicular Transport antagonists & inhibitors, Adaptor Proteins, Vesicular Transport genetics, Extracellular Signal-Regulated MAP Kinases genetics, Extracellular Signal-Regulated MAP Kinases metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, HEK293 Cells, Hep G2 Cells, Heterotrimeric GTP-Binding Proteins antagonists & inhibitors, Heterotrimeric GTP-Binding Proteins genetics, Humans, I-kappa B Kinase genetics, I-kappa B Kinase metabolism, JNK Mitogen-Activated Protein Kinases genetics, JNK Mitogen-Activated Protein Kinases metabolism, Mutation, Phospholipase C beta genetics, Phospholipase C beta metabolism, Phosphorylation, Protein Binding, RNA, Small Interfering genetics, SOS1 Protein genetics, SOS1 Protein metabolism, Son of Sevenless Proteins genetics, Son of Sevenless Proteins metabolism, Transfection, ras Proteins genetics, Adaptor Proteins, Vesicular Transport metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Gene Expression Regulation, Heterotrimeric GTP-Binding Proteins metabolism, Signal Transduction genetics, ras Proteins metabolism
- Abstract
Many G(q) -coupled receptors mediate mitogenic signals by stimulating extracellular signal-regulated protein kinases (ERKs) that are typically regulated by the small GTPase Ras. Recent studies have revealed that members of the Gα(q) family may possess the ability to activate Ras/ERK by interacting with the adaptor protein tetratricopeptide repeat 1 (TPR1). Within the Gα(q) family, the highly promiscuous Gα(14) can relay signals from numerous receptors. Here, we examined if Gα(14) interacts with TPR1 to stimulate Ras signaling pathways. Expression of the constitutively active Gα(14) QL mutant in HEK293 cells led to the formation of GTP-bound Ras as well as increased phosphorylations of downstream signaling molecules including ERK and IκB kinase. Stimulation of endogenous G(14) -coupled somatostatin type 2 and α(2) -adrenergic receptors produced similar responses in human hepatocellular HepG2 carcinoma cells. Co-immunoprecipitation assays using HEK293 cells demonstrated a stronger association of TPR1 for Gα(14) QL than Gα(14) , suggesting that TPR1 preferentially binds to the GTP-bound form of Gα(14) . Activated Gα(14) also interacted with the Ras guanine nucleotide exchange factors SOS1 and SOS2. Expression of a dominant negative mutant of TPR1 or siRNA-mediated knockdown of TPR1 effectively abolished the ability of Gα(14) to induce Ras signaling in native HepG2 or transfected HEK293 cells. Although expression of the dominant negative mutant of TPR1 suppressed Gα(14) QL-induced phosphorylations of ERK and IκB kinase, it did not affect Gα(14) QL-induced stimulation of phospholipase Cβ or c-Jun N-terminal kinase. Our results suggest that TPR1 is required for Gα(14) to stimulate Ras-dependent signaling pathways, but not for the propagation of signals along Ras-independent pathways., (Copyright © 2012 Wiley Periodicals, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
113. The Akt1 isoform is required for optimal IFN-β transcription through direct phosphorylation of β-catenin.
- Author
-
Gantner BN, Jin H, Qian F, Hay N, He B, and Ye RD
- Subjects
- Animals, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Phosphorylation immunology, Protein Isoforms deficiency, Protein Isoforms genetics, Protein Isoforms physiology, Proto-Oncogene Proteins c-akt deficiency, Proto-Oncogene Proteins c-akt genetics, Signal Transduction genetics, Signal Transduction immunology, Toll-Like Receptor 3 metabolism, beta Catenin physiology, Interferon-beta physiology, Proto-Oncogene Proteins c-akt physiology, Transcription, Genetic immunology, beta Catenin metabolism
- Abstract
IFN-β is a critical antiviral cytokine that is capable of modulating the systemic immune response. The transcriptional induction of IFN-β is a highly regulated process, involving the activation of pattern recognition receptors and their downstream signaling pathways. The Akt family of serine/threonine kinases includes three isoforms. The specific role for the individual Akt isoforms in pattern recognition and signaling remains unclear. In this article, we report that the TLR3-mediated expression of IFN-β is blunted in cells that lack Akt1. The expression of IFN-β-inducible genes such as CCL5 and CXCL10 was also reduced in Akt1-deficient cells; the induction of TNF-α and CXCL2, whose expression does not rely on IFN-β, was not reduced in the absence of Akt1. Macrophages from Akt1(-/-) mice displayed deficient clearance of HSV-1 along with reduced IFN-β expression. Our results demonstrate that Akt1 signals through β-catenin by phosphorylation on Ser(552), a site that differs from the glycogen synthase kinase 3 β phosphorylation site. Stimulation of a chemically activated version of Akt1, in the absence of other TLR3-dependent signaling, was sufficient for accumulation and phosphorylation of β-catenin at Ser(552). Taken together, these results demonstrate that the Akt1 isoform is required for β-catenin-mediated promotion of IFN-β transcription downstream of TLR3 activation.
- Published
- 2012
- Full Text
- View/download PDF
114. Protective role of reactive oxygen species in endotoxin-induced lung inflammation through modulation of IL-10 expression.
- Author
-
Deng J, Wang X, Qian F, Vogel S, Xiao L, Ranjan R, Park H, Karpurapu M, Ye RD, Park GY, and Christman JW
- Subjects
- Adoptive Transfer, Animals, Disease Models, Animal, Endotoxins antagonists & inhibitors, Humans, Interleukin-10 antagonists & inhibitors, Luciferases biosynthesis, Luciferases genetics, Lung metabolism, Macrophages, Peritoneal transplantation, Mice, Mice, Knockout, Mice, Transgenic, NADPH Oxidases deficiency, NADPH Oxidases genetics, Pneumonia therapy, Reactive Oxygen Species metabolism, Recombinant Proteins administration & dosage, Endotoxins toxicity, Gene Expression Regulation immunology, Interleukin-10 biosynthesis, Lung immunology, Lung pathology, Pneumonia immunology, Pneumonia pathology, Reactive Oxygen Species therapeutic use
- Abstract
Reactive oxygen species (ROS) generated by NADPH oxidase are generally known to be proinflammatory, and it seems to be counterintuitive that ROS play a critical role in regulating the resolution of the inflammatory response. However, we observed that deficiency of the p47(phox) component of NADPH oxidase in macrophages was associated with a paradoxical accentuation of inflammation in a whole animal model of noninfectious sepsis induced by endotoxin. We have confirmed this observation by interrogating four separate in vivo models that use complementary methodology including the use of p47(phox-/-) mice, p47(phox-/-) bone marrow chimera mice, adoptive transfer of macrophages from p47(phox-/-) mice, and an isolated perfused lung edema model that all point to a relationship between excessive acute inflammation and p47(phox) deficiency in macrophages. Mechanistic data indicate that ROS deficiency in both cells and mice results in decreased production of IL-10 in response to treatment with LPS, at least in part, through attenuation of the Akt-GSK3-β signal pathway and that it can be reversed by the administration of rIL-10. Our data support the innovative concept that generation of ROS is essential for counterregulation of acute lung inflammation.
- Published
- 2012
- Full Text
- View/download PDF
115. Map kinase phosphatase 5 protects against sepsis-induced acute lung injury.
- Author
-
Qian F, Deng J, Gantner BN, Flavell RA, Dong C, Christman JW, and Ye RD
- Subjects
- Acute Lung Injury etiology, Acute Lung Injury immunology, Animals, Bone Marrow Cells immunology, Bone Marrow Cells metabolism, Cells, Cultured, Cytokines biosynthesis, Dual-Specificity Phosphatases deficiency, Dual-Specificity Phosphatases genetics, Escherichia coli, Lipopolysaccharides pharmacology, MAP Kinase Signaling System, Macrophages immunology, Macrophages metabolism, Macrophages physiology, Mice, Mice, Knockout, Mitogen-Activated Protein Kinases metabolism, Nitric Oxide biosynthesis, Phagocytosis, Phosphorylation, Protein Processing, Post-Translational, Sepsis immunology, Superoxides metabolism, Acute Lung Injury enzymology, Dual-Specificity Phosphatases physiology, Sepsis complications
- Abstract
Mitogen-activated protein kinases (MAPKs) play a critical role in inflammation. Although activation of MAPK in inflammatory cells has been studied extensively, much less is known about the inactivation of these kinases. MAPK phosphatase 5 (MKP5) is a member of the dual-specificity phosphatase family that dephosphorylates activated MAPKs. Here we report that MKP5 protects sepsis-induced acute lung injury. Mice lacking MKP5 displayed severe lung tissue damage following LPS challenge, characterized with increased neutrophil infiltration and edema compared with wild-type (WT) controls. In response to LPS, MKP5-deficient macrophages produced significantly more inflammatory factors including inflammatory cytokines, nitric oxide, and superoxide. Phosphorylation of p38 MAPK, JNK, and ERK were enhanced in MKP5-deficient macrophages upon LPS stimulation. Adoptive transfer of MKP5-deficient macrophages led to more severe lung inflammation than transfer of WT macrophages, suggesting that MKP5-deficient macrophages directly contribute to acute lung injury. Taken together, these results suggest that MKP5 is crucial to homeostatic regulation of MAPK activation in inflammatory responses.
- Published
- 2012
- Full Text
- View/download PDF
116. Bidirectional regulation of neutrophil migration by mitogen-activated protein kinases.
- Author
-
Liu X, Ma B, Malik AB, Tang H, Yang T, Sun B, Wang G, Minshall RD, Li Y, Zhao Y, Ye RD, and Xu J
- Subjects
- Animals, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Flavonoids pharmacology, HEK293 Cells, HL-60 Cells, Humans, Imidazoles pharmacology, Mice, Mice, Knockout, N-Formylmethionine Leucyl-Phenylalanine metabolism, Neutrophils drug effects, Neutrophils enzymology, Pyridines pharmacology, Signal Transduction drug effects, Signal Transduction physiology, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, Chemotaxis, Leukocyte, Extracellular Signal-Regulated MAP Kinases metabolism, G-Protein-Coupled Receptor Kinase 2 metabolism, Neutrophils immunology, Receptors, Formyl Peptide metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
To kill invading bacteria, neutrophils must interpret spatial cues, migrate and reach target sites. Although the initiation of chemotactic migration has been extensively studied, little is known about its termination. Here we found that two mitogen-activated protein kinases (MAPKs) had opposing roles in neutrophil trafficking. The extracellular signal-regulated kinase Erk potentiated activity of the G protein-coupled receptor kinase GRK2 and inhibited neutrophil migration, whereas the MAPK p38 acted as a noncanonical GRK that phosphorylated the formyl peptide receptor FPR1 and facilitated neutrophil migration by blocking GRK2 function. Therefore, the dynamic balance between Erk and p38 controlled neutrophil 'stop' and 'go' activity, which ensured that neutrophils reached their final destination as the first line of host defense.
- Published
- 2012
- Full Text
- View/download PDF
117. Identification of a nuclear localization sequence in β-arrestin-1 and its functional implications.
- Author
-
Hoeppner CZ, Cheng N, and Ye RD
- Subjects
- Amino Acid Sequence, Arrestins genetics, Cell Nucleus chemistry, Cell Nucleus genetics, HeLa Cells, Humans, Molecular Sequence Data, Protein Transport, beta-Arrestin 1, beta-Arrestins, Arrestins chemistry, Arrestins metabolism, Cell Nucleus metabolism, Nuclear Localization Signals
- Abstract
A mounting body of evidence suggests that β-arrestin-1 plays important roles in the nucleus, but how β-arrestin-1 enters the nucleus remains unclear because no nuclear import signal has been identified in the β-arrestins. We sought to characterize the cellular localization of wild type β-arrestin-1 and a series of N domain mutants to determine the structural basis and functional implications of β-arrestin-1 nuclear localization. A seven-residue candidate nuclear localization sequence (NLS) was identified based on sequence analysis. Mutation of the NLS led to a loss of β-arrestin-1 nuclear localization in transfected cells. Exogenous expression of wild type β-arrestin-1 enhanced the transcriptional activity of nuclear factor κB (NF-κB) induced by bradykinin, whereas mutation of the NLS reduced this effect by two-thirds relative to wild type controls. Loss of β-arrestin-1 nuclear localization was accompanied by reduced recruitment of the CREB-binding protein and altered post-translational modification profile of p65/RelA. Further mutational analysis identified Lys(157) within the putative NLS as being critical to nuclear localization of β-arrestin-1. Substitution of Lys(157) to Ala led to reduced nuclear localization, decreased promoter binding by p65/RelA and decreased IL-1β gene transcription. These results demonstrate a critical role for β-arrestin-1 nuclear localization in scaffolding and transcriptional regulation.
- Published
- 2012
- Full Text
- View/download PDF
118. Role for the guanine nucleotide exchange factor phosphatidylinositol-3,4,5-trisphosphate-dependent rac exchanger 1 in platelet secretion and aggregation.
- Author
-
Qian F, Le Breton GC, Chen J, Deng J, Christman JW, Wu D, and Ye RD
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Adenosine Triphosphate metabolism, Animals, Bleeding Time, Blood Platelets drug effects, Blotting, Western, Collagen pharmacology, Cytoplasmic Granules metabolism, Dose-Response Relationship, Drug, Enzyme Activation, Female, Genotype, Guanine Nucleotide Exchange Factors deficiency, Guanine Nucleotide Exchange Factors genetics, HEK293 Cells, Hemostasis genetics, Humans, JNK Mitogen-Activated Protein Kinases metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Phosphorylation, Proto-Oncogene Proteins c-akt metabolism, Receptors, G-Protein-Coupled agonists, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Thrombin metabolism, Transfection, p38 Mitogen-Activated Protein Kinases metabolism, rac1 GTP-Binding Protein metabolism, Blood Platelets metabolism, Guanine Nucleotide Exchange Factors metabolism, Platelet Aggregation drug effects
- Abstract
Objective: Recent studies have shown a role for Rac1 in regulating platelet functions, but how Rac1 is activated in platelets remains unclear. Phosphatidylinositol-3,4,5-trisphosphate-dependent Rac exchanger 1 (P-Rex1) was originally identified in neutrophils that regulates phagocyte functions. We sought to examine whether P-Rex1 plays a role in platelet activation., Methods and Results: Western blotting showed P-Rex1 expression in mouse and human platelets. Mice lacking P-Rex1 exhibited prolonged bleeding time and increased rebleeding. When challenged with low doses of the G protein-coupled receptor (GPCR) agonists U46619 and thrombin, P-Rex1-/- platelets displayed significantly reduced secretion and aggregation compared with wild-type platelets. Increasing the concentration of these agonists could overcome the defect. Platelet aggregation induced by collagen, a non-GPCR agonist, was also compromised in the absence of P-Rex1. Along with these phenotypic changes were impaired Rac1 activation; reduced ATP secretion; and decreased phosphorylation of Akt, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase in P-Rex1-/- platelets on agonist stimulation., Conclusion: These results demonstrate for the first time the presence of P-Rex1 in platelets as well as its role in platelet secretion and aggregation induced by low-dose agonists for GPCR and by collagen.
- Published
- 2012
- Full Text
- View/download PDF
119. Role of G protein-coupled receptors in inflammation.
- Author
-
Sun L and Ye RD
- Subjects
- Animals, Humans, Signal Transduction, Inflammation metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
G protein-coupled receptors (GPCRs) play important roles in inflammation. Inflammatory cells such as polymorphonuclear leukocytes (PMN), monocytes and macrophages express a large number of GPCRs for classic chemoattractants and chemokines. These receptors are critical to the migration of phagocytes and their accumulation at sites of inflammation, where these cells can exacerbate inflammation but also contribute to its resolution. Besides chemoattractant GPCRs, protease activated receptors (PARs) such as PAR1 are involved in the regulation of vascular endothelial permeability. Prostaglandin receptors play different roles in inflammatory cell activation, and can mediate both proinflammatory and anti-inflammatory functions. Many GPCRs present in inflammatory cells also mediate transcription factor activation, resulting in the synthesis and secretion of inflammatory factors and, in some cases, molecules that suppress inflammation. An understanding of the signaling paradigms of GPCRs in inflammatory cells is likely to facilitate translational research and development of improved anti-inflammatory therapies.
- Published
- 2012
- Full Text
- View/download PDF
120. The redox-sensitive cation channel TRPM2 modulates phagocyte ROS production and inflammation.
- Author
-
Di A, Gao XP, Qian F, Kawamura T, Han J, Hecquet C, Ye RD, Vogel SM, and Malik AB
- Subjects
- Animals, Calcium metabolism, Inflammation genetics, Inflammation pathology, Lung Diseases genetics, Lung Diseases metabolism, Lung Diseases pathology, Membrane Potentials, Mice, Mice, Inbred C57BL, Mice, Knockout, Oxidation-Reduction, TRPM Cation Channels deficiency, TRPM Cation Channels genetics, Inflammation metabolism, Phagocytes metabolism, Reactive Oxygen Species metabolism, TRPM Cation Channels metabolism
- Abstract
The NADPH oxidase activity of phagocytes and its generation of reactive oxygen species (ROS) is critical for host defense, but ROS overproduction can also lead to inflammation and tissue injury. Here we report that TRPM2, a nonselective and redox-sensitive cation channel, inhibited ROS production in phagocytic cells and prevented endotoxin-induced lung inflammation in mice. TRPM2-deficient mice challenged with endotoxin (lipopolysaccharide) had an enhanced inflammatory response and diminished survival relative to that of wild-type mice challenged with endotoxin. TRPM2 functioned by dampening NADPH oxidase-mediated ROS production through depolarization of the plasma membrane in phagocytes. As ROS also activate TRPM2, our findings establish a negative feedback mechanism for the inactivation of ROS production through inhibition of the membrane potential-sensitive NADPH oxidase.
- Published
- 2011
- Full Text
- View/download PDF
121. Characterization of Quin-C1 for its anti-inflammatory property in a mouse model of bleomycin-induced lung injury.
- Author
-
He M, Cheng N, Gao WW, Zhang M, Zhang YY, Ye RD, and Wang MW
- Subjects
- Animals, Bleomycin toxicity, Chemokines drug effects, Chemokines metabolism, Cytokines drug effects, Cytokines metabolism, Disease Models, Animal, Fibrosis, Inflammation physiopathology, Lung Injury physiopathology, Male, Mice, Mice, Inbred ICR, Receptors, Formyl Peptide agonists, Time Factors, Anti-Inflammatory Agents pharmacology, Benzamides pharmacology, Inflammation drug therapy, Lung Injury drug therapy, Quinazolines pharmacology
- Abstract
Aim: To study the in vivo effects of Quin-C1, a highly specific agonist for formyl peptide receptor 2 (FPR2/ALX), in a mouse model of bleomycin (BLM)-induced lung injury., Methods: Male ICR mice were injected intratracheally with BLM (d 0), and intraperitoneally with Quin-C1 (0.2 mg/d) or vehicle between d 1 and d 28, during which pulmonary inflammation was monitored. A similar regimen was carried out between d 5 and d 28 to differentiate anti-inflammatory from anti-fibrotic effects. During the treatment, leukocyte numbers in bronchoalveolar lavage fluid (BALF) were counted, and FPR2/ALX transcripts, tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), the mouse keratinocyte-derived chemokine (KC), transforming growth factor β1 (TGF-β1) and C-X-C motif chemokine 10 (CXCL10) expression levels in the lung tissue were also measured. Both hydroxyproline content and histological changes were examined on d 28 to assess the severity of lung fibrosis., Results: BLM caused a significant increase in expression levels of all the selected cytokines and chemokines, as well as a thickening of the alveolar wall. Treatment with Quin-C1 significantly reduced the neutrophil and lymphocyte counts in BALF, diminished expression of TNF-α, IL-1β, KC, and TGF-β1, and decreased collagen deposition in lung tissue. The treatment also lowered the content of lung hydroxyproline. Quin-C1 did not ameliorate lung fibrosis when the treatment was started 5 d after the BLM challenge, suggesting that the protection may be attributed to its anti-inflammatory effects. Exposure to BLM or BLM plus Quin-C1 did not change the level of FPR2/ALX transcripts (mFpr1, mFpr2, and Lxa4r) in the lung tissue., Conclusion: The results demonstrate an anti-inflammatory role for Quin-C1 in bleomycin-induced lung injury, which may be further explored for therapeutic applications.
- Published
- 2011
- Full Text
- View/download PDF
122. Gα16 interacts with tetratricopeptide repeat 1 (TPR1) through its β3 region to activate Ras independently of phospholipase Cβ signaling.
- Author
-
Liu AM, Lo RKh, Guo EX, Ho MK, Ye RD, and Wong YH
- Subjects
- Enzyme Activation, HEK293 Cells, Humans, Models, Molecular, Phospholipase C beta metabolism, Protein Binding, Protein Structure, Tertiary, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Signal Transduction, Adaptor Proteins, Vesicular Transport metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 chemistry, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Heterotrimeric GTP-Binding Proteins metabolism, ras Proteins metabolism
- Abstract
Background: G protein-coupled receptors constitute the largest family of cell surface receptors in the mammalian genome. As the core of the G protein signal transduction machinery, the Gα subunits are required to interact with multiple partners. The GTP-bound active state of many Gα subunits can bind a multitude of effectors and regulatory proteins. Yet it remains unclear if the different proteins utilize distinct or common structural motifs on the Gα subunit for binding. Using Gα16 as a model, we asked if its recently discovered adaptor protein tetratricopeptide repeat 1 (TPR1) binds to the same region as its canonical effector, phospholipase Cβ (PLCβ)., Results: We have examined the specificity of Gα16/TPR1 association by testing a series of chimeras between Gα16 and Gαz. TPR1 co-immunoprecipitated with Gα16 and more tightly with its constitutively active Gα16QL, but not Gαz. Progressive replacement of Gα16 sequence with the corresponding residues of Gαz eventually identified a stretch of six amino acids in the β3 region of Gα16 which are responsible for TPR1 interaction and the subsequent Ras activation. Insertion of these six residues into Gαz allowed productive TPR1-interaction. Since the β3 region only minimally contributes to interact with PLCβ, several chimeras exhibited differential abilities to stimulate PLCβ and Ras. The ability of the chimeras to activate downstream transcription factors such as signal transducer and activator of transcription 3 and nuclear factor κB appeared to be associated with PLCβ signaling., Conclusions: Our results suggest that Gα16 can signal through TPR1/Ras and PLCβ simultaneously and independently. The β3 region of Gα16 is essential for interaction with TPR1 and the subsequent activation of Ras, but has relatively minor influence on the PLCβ interaction. Gα16 may utilize different structural domains to bind TPR1 and PLCβ.
- Published
- 2011
- Full Text
- View/download PDF
123. Cell type-specific release of matrix-metallo-proteinase-9 by bacterial chemoattractant in human blood phagocytic leukocytes.
- Author
-
Doerner AM, Chen LY, Ye RD, Yong J, Huang S, and Pan ZK
- Abstract
Stimulation of phagocytic leukocytes with bacterial chemoattractant resulted in the release of matrix metal-loproteinases (MMPs). Little is known about the mechanisms of bacterial chemoattractant regulation of MMP in phagocytic leukocytes. We report here that the mechanisms of the bacterial chemotactic peptidefMLP-induced MMP -9 release in monocytes appeared to be different from fMLP-stimulated MMP-9 release in neutrophils. In freshly prepared peripheral blood monocytes, fMLP induces MMP-9 release, starting at 8 h after stimulation. These functions of fMLP is accompanied by an increase in TNFα expression, and mediated through the phosphorylation of ERK1/2 in monocytes. However, neutrophil preparations that responded to fMLP with MMP-9 release did not require activation of ERK1/2 and TNFα expression. These results suggest a different role of fMLP in MMP-9 expression in neutrophils and monocytes, and the signal molecules involved in mediating this effect in human blood monocytes stimulated by bacterial chemoattractant.
- Published
- 2011
124. Polymerization of MIP-1 chemokine (CCL3 and CCL4) and clearance of MIP-1 by insulin-degrading enzyme.
- Author
-
Ren M, Guo Q, Guo L, Lenz M, Qian F, Koenen RR, Xu H, Schilling AB, Weber C, Ye RD, Dinner AR, and Tang WJ
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Chemokine CCL3 genetics, Chemokine CCL3 immunology, Chemokine CCL4 genetics, Chemokine CCL4 immunology, Crystallography, X-Ray, Humans, Insulysin chemistry, Macrophage Inflammatory Proteins chemistry, Macrophage Inflammatory Proteins genetics, Macrophage Inflammatory Proteins immunology, Macrophage Inflammatory Proteins metabolism, Male, Mice, Mice, Inbred C57BL, Models, Molecular, Molecular Sequence Data, Mutation, Polymerization, Protein Binding, Protein Conformation, Protein Multimerization, Chemokine CCL3 chemistry, Chemokine CCL3 metabolism, Chemokine CCL4 chemistry, Chemokine CCL4 metabolism, Insulysin metabolism
- Abstract
Macrophage inflammatory protein-1 (MIP-1), MIP-1α (CCL3) and MIP-1β (CCL4) are chemokines crucial for immune responses towards infection and inflammation. Both MIP-1α and MIP-1β form high-molecular-weight aggregates. Our crystal structures reveal that MIP-1 aggregation is a polymerization process and human MIP-1α and MIP-1β form rod-shaped, double-helical polymers. Biophysical analyses and mathematical modelling show that MIP-1 reversibly forms a polydisperse distribution of rod-shaped polymers in solution. Polymerization buries receptor-binding sites of MIP-1α, thus depolymerization mutations enhance MIP-1α to arrest monocytes onto activated human endothelium. However, same depolymerization mutations render MIP-1α ineffective in mouse peritoneal cell recruitment. Mathematical modelling reveals that, for a long-range chemotaxis of MIP-1, polymerization could protect MIP-1 from proteases that selectively degrade monomeric MIP-1. Insulin-degrading enzyme (IDE) is identified as such a protease and decreased expression of IDE leads to elevated MIP-1 levels in microglial cells. Our structural and proteomic studies offer a molecular basis for selective degradation of MIP-1. The regulated MIP-1 polymerization and selective inactivation of MIP-1 monomers by IDE could aid in controlling the MIP-1 chemotactic gradient for immune surveillance.
- Published
- 2010
- Full Text
- View/download PDF
125. Galpha16 activates Ras by forming a complex with tetratricopeptide repeat 1 (TPR1) and Son of Sevenless (SOS).
- Author
-
Liu AM, Lo RK, Lee MM, Wang Y, Yeung WW, Ho MK, Su Y, Ye RD, and Wong YH
- Subjects
- Cell Line, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, Humans, Mutation, Phospholipase C beta metabolism, Signal Transduction, ras GTPase-Activating Proteins metabolism, Adaptor Proteins, Vesicular Transport metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Heterotrimeric GTP-Binding Proteins metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Son of Sevenless Proteins metabolism
- Abstract
Many G protein-coupled receptors (GPCRs) are known to modulate cell growth and differentiation by stimulating the extracellular signal-regulated protein kinases (ERKs). In growth factor signaling, ERKs are typically stimulated through an elaborate network of modules consisting of adaptors, protein kinases, and the small GTPase Ras. The mechanism by which G protein signals tap into the ERK signaling pathway has thus far remain elusive. Members of the Gq family of G proteins, in particular Galpha16, have been shown to associate with tetratricopeptide repeat 1 (TPR1), an adaptor protein which preferentially binds to Ras. Here, we examined if TPR1 is indeed the missing link between Galpha16 signaling and Ras activation. Expression of Galpha16QL, a constitutively active mutant of Galpha16, in HEK 293 cells led to the formation of GTP-bound Ras and the subsequent phosphorylation of ERK. Likewise, stimulation of endogenou G16-coupled CCR1 chemokine receptors produced the same responses in human erythroleukemia cells. siRNA-mediated knockdown of TPR1 or expression of a dominant negative mutant of TPR1 effectively abolished the ability of Galpha16QL to induce Ras activation in HEK 293 cells. In contrast, these manipulations had no inhibitory effect on Galpha16QL induced activation of phospholipase Cbeta. Galpha16QL-induced phosphorylations of downstream targets including ERK, signal transducer and activator of transcription 3, and IkappaB kinase were significantly suppressed upon expression of the dominant negative mutant of TPR1. Furthermore, SOS2, a Ras guanine nucleotide exchange factor, was found to form a complex with TPR1 and Galpha16QL. Expression of SOS2 enhanced Galpha16QL-induced Ras activation and its subsequent signaling. Collectively, our results suggest that Galpha16 regulates multiple signaling pathways by activating Ras through its association with TPR1, but TPR1 is not required for Galpha16 to stimulate phospholipase Cbeta.
- Published
- 2010
- Full Text
- View/download PDF
126. Heterotrimeric G protein signaling outside the realm of seven transmembrane domain receptors.
- Author
-
Marty C and Ye RD
- Subjects
- Animals, Heterotrimeric GTP-Binding Proteins drug effects, Heterotrimeric GTP-Binding Proteins physiology, Humans, Receptor Protein-Tyrosine Kinases metabolism, Heterotrimeric GTP-Binding Proteins metabolism, Signal Transduction
- Abstract
Heterotrimeric G proteins, consisting of the guanine nucleotide-binding Galpha subunits with GTPase activity and the closely associated Gbeta and Ggamma subunits, are important signaling components for receptors with seven transmembrane domains (7TMRs). These receptors, also termed G protein-coupled receptors (GPCRs), act as guanine nucleotide exchange factors upon agonist stimulation. There is now accumulating evidence for noncanonical functions of heterotrimeric G proteins independent of 7TMR coupling. Galpha proteins belonging to all 4 subfamilies, including G(s), G(i), G(q), and G(12) are found to play important roles in receptor tyrosine kinase signaling, regulation of oxidant production, development, and cell migration, through physical and functional interaction with proteins other than 7TMRs. Association of Galpha with non-7TMR proteins also facilitates presentation of these G proteins to specific cellular microdomains. This Minireview aims to summarize our current understanding of the noncanonical roles of Galpha proteins in cell signaling and to discuss unresolved issues including regulation of Galpha activation by proteins other than the 7TMRs.
- Published
- 2010
- Full Text
- View/download PDF
127. Editorial: Biased agonism in chemoattractant receptor signaling.
- Author
-
Ye RD
- Subjects
- Animals, Humans, Ligands, Receptor, Anaphylatoxin C5a genetics, Respiratory Burst, Chemotactic Factors metabolism, Chemotaxis, Leukocyte physiology, DNA-Binding Proteins metabolism, Escherichia coli Proteins metabolism, Molecular Chaperones metabolism, Neutrophils metabolism, Receptor, Anaphylatoxin C5a metabolism
- Published
- 2010
- Full Text
- View/download PDF
128. Akt isoforms differentially regulate neutrophil functions.
- Author
-
Chen J, Tang H, Hay N, Xu J, and Ye RD
- Subjects
- Animals, Biological Transport, Active, Cell Degranulation, Cell Movement, In Vitro Techniques, Mice, Mice, Inbred C57BL, Mice, Knockout, NADPH Oxidases metabolism, Neutrophil Activation drug effects, Neutrophil Activation physiology, Neutrophils drug effects, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Proto-Oncogene Proteins c-akt deficiency, Proto-Oncogene Proteins c-akt genetics, Signal Transduction, Superoxides metabolism, Tetradecanoylphorbol Acetate pharmacology, Neutrophils physiology, Proto-Oncogene Proteins c-akt physiology
- Abstract
In neutrophils, the phosphoinositide 3-kinase/Akt signaling cascade is involved in migration, degranulation, and O(2)(-) production. However, it is unclear whether the Akt kinase isoforms have distinct functions in neutrophil activation. Here we report functional differences between the 2 major Akt isoforms in neutrophil activation on the basis of studies in which we used individual Akt1 and Akt2 knockout mice. Akt2(-/-) neutrophils exhibited decreased cell migration, granule enzyme release, and O(2)(-) production compared with wild-type and Akt1(-/-) neutrophils. Surprisingly, Akt2 deficiency and pharmacologic inhibition of Akt also abrogated phorbol ester-induced O(2)(-) production, which was unaffected by treatment with the phosphoinositide 3-kinase inhibitor LY294002. The decreased O(2)(-) production in Akt2(-/-) neutrophils was accompanied by reduced p47(phox) phosphorylation and its membrane translocation, suggesting that Akt2 is important for the assembly of phagocyte nicotinamide adenine dinucleotide phosphate oxidase. In wild-type neutrophils, Akt2 but not Akt1 translocated to plasma membrane upon chemoattractant stimulation and to the leading edge in polarized neutrophils. In the absence of Akt2, chemoattractant-induced Akt protein phosphorylation was significantly reduced. These results demonstrate a predominant role of Akt2 in regulating neutrophil functions and provide evidence for differential activation of the 2 Akt isoforms in neutrophils.
- Published
- 2010
- Full Text
- View/download PDF
129. Characterization of P-Rex1 for its role in fMet-Leu-Phe-induced superoxide production in reconstituted COS(phox) cells.
- Author
-
Nie B, Cheng N, Dinauer MC, and Ye RD
- Subjects
- Amino Acid Substitution genetics, Animals, COS Cells, Cell Membrane drug effects, Cell Membrane enzymology, Chlorocebus aethiops, Enzyme Activation drug effects, Feedback, Physiological drug effects, GTP-Binding Protein beta Subunits metabolism, GTP-Binding Protein gamma Subunits metabolism, Guanine Nucleotide Exchange Factors chemistry, Humans, Mice, Models, Biological, Mutation genetics, Phosphatidylinositol 3-Kinases metabolism, Protein Kinase C-delta metabolism, Protein Structure, Tertiary, Proto-Oncogene Proteins c-akt metabolism, rac1 GTP-Binding Protein metabolism, Guanine Nucleotide Exchange Factors metabolism, N-Formylmethionine Leucyl-Phenylalanine pharmacology, NADPH Oxidases metabolism, Superoxides metabolism
- Abstract
P-Rex1 (phosphatidylinositol 3,4,5-trisphosphate-dependent Rac exchanger 1) is a Rac-specific guanine nucleotide exchange factor activated by Gbetagamma subunits and by PtdIns((3,4,5))P(3). Recent studies indicate that P-Rex1 plays an important role in signaling downstream of neutrophil chemoattractant receptors. Here we report that heterologous expression of P-Rex1, but not Vav1, reconstitutes formyl peptide receptor 1 (FPR1)-mediated NADPH oxidase activation in the transgenic COS(phox) cells expressing gp91(phox), p22(phox), p67(phox) and p47(phox). A successful reconstitution requires the expression of a full-length P-Rex1 with intact DH and PH domains, and is accompanied by P-Rex1 membrane localization as well as Rac1 activation. P-Rex1-dependent superoxide generation in the reconstituted COS(phox) cells was further enhanced by expression of the novel PKC isoform PKCdelta and by overexpression of Akt. Heterologous expression of P-Rex1 in COS(phox) cells potentiated fMet-Leu-Phe-induced Akt phosphorylation, whereas expression of a constitutively active form of Akt enhanced Rac1 activation. In contrast, a dominant negative Akt mutant reduced the fMet-Leu-Phe stimulated superoxide generation as well as Rac1 activation. These results demonstrate that in COS(phox) cells, P-Rex1 is a critical component for FPR1-mediated signaling leading to NADPH oxidase activation, and there is a crosstalk between the P-Rex1-Rac pathway and Akt in superoxide generation., (2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
130. Identification of novel small-molecule agonists for human formyl peptide receptors and pharmacophore models of their recognition.
- Author
-
Kirpotina LN, Khlebnikov AI, Schepetkin IA, Ye RD, Rabiet MJ, Jutila MA, and Quinn MT
- Subjects
- 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, Models, Molecular, Receptors, Formyl Peptide agonists, Receptors, Formyl Peptide metabolism, Small Molecule Libraries metabolism
- 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.
- Published
- 2010
- Full Text
- View/download PDF
131. A non-redundant role for MKP5 in limiting ROS production and preventing LPS-induced vascular injury.
- Author
-
Qian F, Deng J, Cheng N, Welch EJ, Zhang Y, Malik AB, Flavell RA, Dong C, and Ye RD
- Subjects
- Animals, Dual-Specificity Phosphatases genetics, Gene Deletion, Mice, Microvessels immunology, NADPH Oxidases metabolism, Neutrophils immunology, Neutrophils metabolism, Reactive Oxygen Species metabolism, Superoxides metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Dual-Specificity Phosphatases metabolism, Lipopolysaccharides immunology, Microvessels injuries
- Abstract
There are at least 11 mitogen-activated protein kinase (MAPK) phosphatases (MKPs) and only 3 major groups of MAPKs, raising the question of whether these phosphatases have non-redundant functions in vivo. Using a modified mouse model of local Shwartzman reaction, we found that deletion of the MKP5 gene, but not the MKP1 gene, led to robust and accelerated vascular inflammatory responses to a single dose of LPS injection. Depletion of neutrophils significantly reduced the vascular injury in Mkp5(-/-) mice, whereas adoptive transfer of Mkp5(-/-) neutrophils replicated the LPS-induced skin lesions in wild-type recipients. Neutrophils isolated from Mkp5(-/-) mice exhibited augmented p38 MAPK activation and increased superoxide generation on activation. The p38 MAPK inhibitor, SB203580, significantly reduced p47(phox) phosphorylation and diminished superoxide production in neutrophils. p38 MAPK phosphorylated mouse p47(phox), and deletion of the p47(phox) gene ablated the LPS-induced vascular injury in Mkp5(-/-) mice. Collectively, these results show an earlier unrecognized and non-redundant function of MKP5 in restraining p38 MAPK-mediated neutrophil oxidant production, thereby preventing LPS-induced vascular injury.
- Published
- 2009
- Full Text
- View/download PDF
132. LIM kinase 1 promotes endothelial barrier disruption and neutrophil infiltration in mouse lungs.
- Author
-
Gorovoy M, Han J, Pan H, Welch E, Neamu R, Jia Z, Predescu D, Vogel S, Minshall RD, Ye RD, Malik AB, and Voyno-Yasenetskaya T
- Subjects
- Acute Lung Injury chemically induced, Acute Lung Injury genetics, Animals, Chemotaxis drug effects, Endothelium pathology, Humans, Lim Kinases genetics, Lipopolysaccharides toxicity, Lung enzymology, Lung pathology, Mice, Mice, Knockout, Neutrophils pathology, Pulmonary Edema chemically induced, Pulmonary Edema enzymology, Pulmonary Edema genetics, Sepsis chemically induced, Sepsis genetics, rho GTP-Binding Proteins genetics, rho GTP-Binding Proteins metabolism, rho-Associated Kinases metabolism, rhoA GTP-Binding Protein, Acute Lung Injury enzymology, Endothelium enzymology, Lim Kinases metabolism, Neutrophil Infiltration, Neutrophils enzymology, Sepsis enzymology
- Abstract
Rationale: Disruption of endothelial barrier function and neutrophil-mediated injury are two major mechanisms underlying the pathophysiology of sepsis-induced acute lung injury (ALI). Recently we reported that endotoxin induced activation of RhoA in mice lungs that led to the disruption of endothelial barrier and lung edema formation; however, the molecular mechanism of this phenomenon remained unknown., Objective: We reasoned that LIMK1, which participates in the regulation of endothelial cell contractility and is activated by RhoA/Rho kinase pathway, could mediate RhoA-dependent disruption of endothelial barrier function in mouse lungs during ALI. And if that is the case, then attenuation of endothelial cell contractility by downregulating LIMK1 may lead to the enhancement of endothelial barrier function, which could protect mice from endotoxin-induced ALI., Methods and Results: Here we report that LIMK1 deficiency in mice significantly reduced mortality induced by endotoxin. Data showed that lung edema formation, lung microvascular permeability, and neutrophil infiltration into the lungs were suppressed in limk1(-/-) mice., Conclusions: We identified that improvement of endothelial barrier function along with impaired neutrophil chemotaxis were the underlying mechanisms that reduced severity of ALI in limk1(-/-) mice, pointing to a new therapeutic target for diseases associated with acute inflammation of the lungs.
- Published
- 2009
- Full Text
- View/download PDF
133. 6-methyl-2,4-disubstituted pyridazin-3(2H)-ones: a novel class of small-molecule agonists for formyl peptide receptors.
- Author
-
Cilibrizzi A, Quinn MT, Kirpotina LN, Schepetkin IA, Holderness J, Ye RD, Rabiet MJ, Biancalani C, Cesari N, Graziano A, Vergelli C, Pieretti S, Dal Piaz V, and Giovannoni MP
- Subjects
- 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, Pyridazines chemical synthesis, Receptors, Formyl Peptide agonists, Receptors, Lipoxin agonists
- 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.
- Published
- 2009
- Full Text
- View/download PDF
134. Lipopolysaccharide stimulates platelet secretion and potentiates platelet aggregation via TLR4/MyD88 and the cGMP-dependent protein kinase pathway.
- Author
-
Zhang G, Han J, Welch EJ, Ye RD, Voyno-Yasenetskaya TA, Malik AB, Du X, and Li Z
- Subjects
- Animals, Cyclic GMP metabolism, Humans, Lipopolysaccharide Receptors metabolism, Lymphocyte Antigen 96 metabolism, Mice, Platelet Aggregation drug effects, Protein Binding, Thrombin metabolism, Blood Platelets drug effects, Blood Platelets metabolism, Cyclic GMP-Dependent Protein Kinases metabolism, Lipopolysaccharides pharmacology, Myeloid Differentiation Factor 88 metabolism, Signal Transduction drug effects, Toll-Like Receptor 4 metabolism
- Abstract
Bacterial LPS induces rapid thrombocytopenia, hypotension, and sepsis. Although growing evidence suggests that platelet activation plays a critical role in LPS-induced thrombocytopenia and tissue damage, the mechanism of LPS-mediated platelet activation is unclear. In this study, we show that LPS stimulates platelet secretion of dense and alpha granules as indicated by ATP release and P-selectin expression, and thus enhances platelet activation induced by low concentrations of platelet agonists. Platelets express components of the LPS receptor-signaling complex, including TLR (TLR4), CD14, MD2, and MyD88, and the effect of LPS on platelet activation was abolished by an anti-TLR4-blocking Ab or TLR4 knockout, suggesting that the effect of LPS on platelet aggregation requires the TLR4 pathway. Furthermore, LPS-potentiated thrombin- and collagen-induced platelet aggregation and FeCl(3)-induced thrombus formation were abolished in MyD88 knockout mice. LPS also induced cGMP elevation and the stimulatory effect of LPS on platelet aggregation was abolished by inhibitors of NO synthase and the cGMP-dependent protein kinase (PKG). LPS-induced cGMP elevation was inhibited by an anti-TLR4 Ab or by TLR4 deficiency, suggesting that activation of the cGMP/protein kinase G pathway by LPS involves the TLR4 pathway. Taken together, our data indicate that LPS stimulates platelet secretion and potentiates platelet aggregation through a TLR4/MyD88- and cGMP/PKG-dependent pathway.
- Published
- 2009
- Full Text
- View/download PDF
135. International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family.
- Author
-
Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, and Murphy PM
- Subjects
- Amino Acid Sequence, Animals, Humans, Molecular Sequence Data, Pharmacology, Clinical organization & administration, Receptors, Formyl Peptide genetics, Receptors, Formyl Peptide physiology, Internationality, Multigene Family, Pharmacology, Clinical standards, Receptors, Formyl Peptide chemistry, Receptors, Formyl Peptide classification, Terminology as Topic
- Abstract
Formyl peptide receptors (FPRs) are a small group of seven-transmembrane domain, G protein-coupled receptors that are expressed mainly by mammalian phagocytic leukocytes and are known to be important in host defense and inflammation. The three human FPRs (FPR1, FPR2/ALX, and FPR3) share significant sequence homology and are encoded by clustered genes. Collectively, these receptors bind an extraordinarily numerous and structurally diverse group of agonistic ligands, including N-formyl and nonformyl peptides of different composition, that chemoattract and activate phagocytes. N-formyl peptides, which are encoded in nature only by bacterial and mitochondrial genes and result from obligatory initiation of bacterial and mitochondrial protein synthesis with N-formylmethionine, is the only ligand class common to all three human receptors. Surprisingly, the endogenous anti-inflammatory peptide annexin 1 and its N-terminal fragments also bind human FPR1 and FPR2/ALX, and the anti-inflammatory eicosanoid lipoxin A4 is an agonist at FPR2/ALX. In comparison, fewer agonists have been identified for FPR3, the third member in this receptor family. Structural and functional studies of the FPRs have produced important information for understanding the general pharmacological principles governing all leukocyte chemoattractant receptors. This article aims to provide an overview of the discovery and pharmacological characterization of FPRs, to introduce an International Union of Basic and Clinical Pharmacology (IUPHAR)-recommended nomenclature, and to discuss unmet challenges, including the mechanisms used by these receptors to bind diverse ligands and mediate different biological functions.
- Published
- 2009
- Full Text
- View/download PDF
136. Duplex high-throughput flow cytometry screen identifies two novel formylpeptide receptor family probes.
- Author
-
Young SM, Bologa CM, Fara D, Bryant BK, Strouse JJ, Arterburn JB, Ye RD, Oprea TI, Prossnitz ER, Sklar LA, and Edwards BS
- Subjects
- Animals, Cells, Cultured, Chemotactic Factors metabolism, Fluorescent Dyes, Humans, Ligands, Molecular Probes analysis, Rats, Receptors, Formyl Peptide metabolism, Receptors, Lipoxin metabolism, Sensitivity and Specificity, U937 Cells, Flow Cytometry methods, Receptors, Formyl Peptide antagonists & inhibitors, Receptors, Lipoxin antagonists & inhibitors
- Abstract
Of recent, clinical interest have been two related human G-protein coupled receptors: formylpeptide receptor (FPR), linked to antibacterial inflammation and malignant glioma cell metastasis; and FPR like-1 (FPRL1), linked to chronic inflammation in systemic amyloidosis, Alzheimer's disease, and prion diseases. In association with the National Institutes of Health (NIH) Molecular Library Screening Network, we implemented a flow-cytometry-based high-throughput screening (HTS) approach for identifying selective small molecule FPR and FPRL1 ligands. The screening assay measured the ability of test compounds to competitively displace a high-affinity, fluorescein- labeled peptide ligand from FPR, FPRL1, or both. U937 cells expressing FPR and rat basophil leukemia (RBL) cells expressing FPRL1 were tested together in a "duplex" format. The U937 cells were color coded with red-fluorescent dye allowing their distinction during analysis. Compounds, cells, and fluorescent ligand were sequentially combined (no wash) in 15 microl assay volumes in 384-well plates. Throughput averaged approximately 11 min per plate to analyze approximately 4,000 cells ( approximately 2,000/receptor) in a 2 microl aspirate from each well. In primary single concentration HTS of 24,304 NIH Small Molecule Repository compounds, 253 resulted in inhibition >30% (181 for FPR, 72 for FPRL1) of which 40 had selective binding inhibition constants (K(i)) < or = 4 microM (34 for FPR and 6 for FPRL1). An additional 1,446 candidate compounds were selected by structure-activity-relationship analysis of the hits and screened to identify novel ligands for FPR (3570-0208, K(i) = 95 +/- 10 nM) and FPRL1 (BB-V-115, K(i) = 270 +/- 51 nM). Each was a selective antagonist in calcium response assays and the most potent small molecule antagonist reported for its respective receptor to date. The duplex assay format reduced assay time, minimized reagent requirements, and provided selectivity information at every screening stage, thus proving to be an efficient means to screen for selective receptor ligand probes., (2008 International Society for Advancement of Cytometry.)
- Published
- 2009
- Full Text
- View/download PDF
137. A novel fluorescent cross-reactive formylpeptide receptor/formylpeptide receptor-like 1 hexapeptide ligand.
- Author
-
Strouse JJ, Young SM, Mitchell HD, Ye RD, Prossnitz ER, Sklar LA, and Edwards BS
- Subjects
- Animals, Cells, Cultured, Flow Cytometry, Fluoresceins chemistry, Fluorescent Dyes chemistry, Humans, Ligands, Oligopeptides chemistry, Peptides chemistry, Rats, Transfection, U937 Cells, Chemotactic Factors metabolism, Fluoresceins metabolism, Fluorescent Dyes metabolism, Oligopeptides metabolism, Peptides metabolism, Receptors, Formyl Peptide metabolism, Receptors, Lipoxin metabolism
- Abstract
Formylpeptide receptors (FPRs) are implicated in a variety of immunological and inflammatory response cascades. Further understanding of FPR-family ligand interactions could play an integral role in biological and therapeutic discovery. Fluorescent reporter ligands for the family are desirable experimental tools for increased understanding of ligand/receptor interactions. The ligand binding affinity and fluorescent reporting activity of the peptide WK(FL)YMVm was explored though use of the high throughput HyperCyt flow cytometric platform. Relative binding affinities of several known FPR and FPRL1 peptide ligands were compared in a duplex assay format. The fluorescent W-peptide ligand, WK(FL)YMVm, proved to be a high-affinity, cross-reactive reporter ligand for the FPR/FPRL1 duplex assay. Ligand specificity was demonstrated for each receptor, with known, selective peptide ligands. The binding site specificity of the reporter ligand was further verified by a fluorescent confocal microscopy internalization experiment. The fluorescent peptide ligand WK(FL)YMVm binds with high affinity to both FPR and FPRL1. The differential affinities of known peptide ligands were observed with the use of this fluorescent probe in high throughput screening flow cytometry., (2008 International Society for Advancement of Cytometry.)
- Published
- 2009
- Full Text
- View/download PDF
138. Serum amyloid A induces G-CSF expression and neutrophilia via Toll-like receptor 2.
- Author
-
He RL, Zhou J, Hanson CZ, Chen J, Cheng N, and Ye RD
- Subjects
- Acute-Phase Reaction genetics, Acute-Phase Reaction metabolism, Animals, Anti-Bacterial Agents pharmacology, Antibodies pharmacology, Endopeptidase K pharmacology, Gene Expression Regulation drug effects, Granulocyte Colony-Stimulating Factor genetics, Humans, Mice, Mice, Knockout, NF-kappa B pharmacology, Polymyxin B pharmacology, RNA, Messenger biosynthesis, RNA, Messenger genetics, Response Elements physiology, Serum Amyloid A Protein, Toll-Like Receptor 2 genetics, Gene Expression Regulation physiology, Granulocyte Colony-Stimulating Factor biosynthesis, Inflammation Mediators metabolism, Macrophages metabolism, Toll-Like Receptor 2 metabolism
- Abstract
The acute-phase protein serum amyloid A (SAA) is commonly considered a marker for inflammatory diseases; however, its precise role in inflammation and infection, which often result in neutrophilia, remains ambiguous. In this study, we demonstrate that SAA is a potent endogenous stimulator of granulocyte colony-stimulated factor (G-CSF), a principal cytokine-regulating granulocytosis. This effect of SAA is dependent on Toll-like receptor 2 (TLR2). Our data demonstrate that, in mouse macrophages, both G-CSF mRNA and protein were significantly increased after SAA stimulation. The induction of G-CSF was blocked by an anti-TLR2 antibody and markedly decreased in the TLR2-deficient macrophages. SAA stimulation results in the activation of nuclear factor-kappaB and binding activity to the CK-1 element of the G-CSF promoter region. In vitro reconstitution experiments also support that TLR2 mediates SAA-induced G-CSF expression. In addition, SAA-induced secretion of G-CSF was sensitive to heat and proteinase K treatment, yet insensitive to polymyxin B treatment, indicating that the induction is a direct effect of SAA. Finally, our in vivo studies confirmed that SAA treatment results in a significant increase in plasma G-CSF and neutrophilia, whereas these responses are ablated in G-CSF- or TLR2-deficient mice.
- Published
- 2009
- Full Text
- View/download PDF
139. Ca2+ entry via TRPC channels is necessary for thrombin-induced NF-kappaB activation in endothelial cells through AMP-activated protein kinase and protein kinase Cdelta.
- Author
-
Bair AM, Thippegowda PB, Freichel M, Cheng N, Ye RD, Vogel SM, Yu Y, Flockerzi V, Malik AB, and Tiruppathi C
- Subjects
- AMP-Activated Protein Kinases genetics, Animals, Calcium, Calcium-Calmodulin-Dependent Protein Kinases genetics, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cells, Cultured, Endothelial Cells cytology, Enzyme Activation drug effects, Humans, Mice, Mice, Knockout, Protein Kinase C-delta genetics, Signal Transduction drug effects, Signal Transduction genetics, TRPC Cation Channels genetics, Thrombosis genetics, Thrombosis metabolism, Transcription Factor RelA genetics, AMP-Activated Protein Kinases metabolism, Endothelial Cells metabolism, Hemostatics pharmacology, Protein Kinase C-delta metabolism, TRPC Cation Channels metabolism, Thrombin pharmacology, Transcription Factor RelA metabolism
- Abstract
The transient receptor potential canonical (TRPC) family channels are proposed to be essential for store-operated Ca2+ entry in endothelial cells. Ca2+ signaling is involved in NF-kappaB activation, but the role of store-operated Ca2+ entry is unclear. Here we show that thrombin-induced Ca2+ entry and the resultant AMP-activated protein kinase (AMPK) activation targets the Ca2+-independent protein kinase Cdelta (PKCdelta) to mediate NF-kappaB activation in endothelial cells. We observed that thrombin-induced p65/RelA, AMPK, and PKCdelta activation were markedly reduced by knockdown of the TRPC isoform TRPC1 expressed in human endothelial cells and in endothelial cells obtained from Trpc4 knock-out mice. Inhibition of Ca2+/calmodulin-dependent protein kinase kinase beta downstream of the Ca2+ influx or knockdown of the downstream Ca2+/calmodulin-dependent protein kinase kinase beta target kinase, AMPK, also prevented NF-kappaB activation. Further, we observed that AMPK interacted with PKCdelta and phosphorylated Thr505 in the activation loop of PKCdelta in thrombin-stimulated endothelial cells. Expression of a PKCdelta-T505A mutant suppressed the thrombin-induced but not the TNF-alpha-induced NF-kappaB activation. These findings demonstrate a novel mechanism for TRPC channels to mediate NF-kappaB activation in endothelial cells that involves the convergence of the TRPC-regulated signaling at AMPK and PKCdelta and that may be a target of interference of the inappropriate activation of NF-kappaB associated with thrombosis.
- Published
- 2009
- Full Text
- View/download PDF
140. beta-Arrestin1 interacts with the G-protein subunits beta1gamma2 and promotes beta1gamma2-dependent Akt signalling for NF-kappaB activation.
- Author
-
Yang M, He RL, Benovic JL, and Ye RD
- Subjects
- Arrestins genetics, Blotting, Western, Cell Line, Electrophoretic Mobility Shift Assay, HeLa Cells, Humans, Immunoprecipitation, Phosphorylation, Protein Binding, RNA, Small Interfering genetics, Sequence Deletion, Signal Transduction, beta-Arrestins, Arrestins metabolism, GTP-Binding Protein beta Subunits metabolism, GTP-Binding Proteins metabolism, NF-kappa B metabolism, Proto-Oncogene Proteins c-akt metabolism
- Abstract
beta-Arrestins are known to regulate G-protein signalling through interactions with their downstream effectors. In the present study, we report that beta-arrestin1 associates with the G-protein beta1gamma2 subunits in transfected cells, and purified beta-arrestin1 interacts with G(beta1gamma2) derived from in vitro translation. Deletion mutagenesis of beta-arrestin1 led to the identification of a region, comprising amino acids 181-280, as being responsible for its interaction with G(beta1gamma2). Overexpression of beta-arrestin1 facilitates G(beta1gamma2)-mediated Akt phosphorylation, and inhibition of endogenous beta-arrestin1 expression by siRNA (small interfering RNA) diminishes this effect. Through investigation of NF-kappaB (nuclear factor kappaB), a transcription factor regulated by Akt signalling, we have found that overexpression of beta-arrestin1 significantly enhances G(beta1gamma2)-mediated nuclear translocation of NF-kappaB proteins and expression of a NF-kappaB-directed luciferase reporter. Overexpression of beta-arrestin1 also promotes bradykinin-induced, G(betagamma)-mediated NF-kappaB luciferase-reporter expression, which is reverted by silencing the endogenous beta-arrestin1 with a specific siRNA. These results identify novel functions of beta-arrestin1 in binding to the beta1gamma2 subunits of heterotrimeric G-proteins and promoting G(betagamma)-mediated Akt signalling for NF-kappaB activation.
- Published
- 2009
- Full Text
- View/download PDF
141. High-content screening: flow cytometry analysis.
- Author
-
Edwards BS, Young SM, Ivnitsky-Steele I, Ye RD, Prossnitz ER, and Sklar LA
- Subjects
- Animals, Binding, Competitive, Humans, Ligands, Rats, Receptors, Formyl Peptide genetics, Receptors, Lipoxin genetics, Tumor Cells, Cultured, Flow Cytometry methods, Leukemia, Basophilic, Acute metabolism, Receptors, Formyl Peptide metabolism, Receptors, Lipoxin metabolism
- Abstract
The HyperCyt high-throughput (HT) flow cytometry sampling platform uses a peristaltic pump, in combination with an autosampler, and a novel approach to data collection, to circumvent time-delay bottlenecks of conventional flow cytometry. This approach also dramatically reduces the amount of sample aspirated for each analysis, typically requiring ~2 microL per sample while making quantitative fluorescence measurements of 40 or more samples per minute with thousands to tens of thousands of cells in each sample. Here, we describe a simple robust screening assay that exploits the high-content measurement capabilities of the flow cytometer to simicroltaneously probe the binding of test compounds to two different receptors in a common assay volume, a duplex assay format. The ability of the flow cytometer to distinguish cell-bound from free fluorophore is also exploited to eliminate wash steps during assay setup. HT flow cytometry with this assay has allowed efficient screening of tens of thousands of small molecules from the NIH Small-Molecule Repository to identify selective ligands for two related G-protein-coupled receptors, the formylpeptide receptor and formylpeptide receptor-like 1.
- Published
- 2009
- Full Text
- View/download PDF
142. Opposing effects of platelet-activating factor and lyso-platelet-activating factor on neutrophil and platelet activation.
- Author
-
Welch EJ, Naikawadi RP, Li Z, Lin P, Ishii S, Shimizu T, Tiruppathi C, Du X, Subbaiah PV, and Ye RD
- Subjects
- Animals, Blood Platelets metabolism, Humans, Mice, Mice, Inbred C57BL, Neutrophils metabolism, Platelet Activating Factor antagonists & inhibitors, Blood Platelets drug effects, Neutrophil Activation drug effects, Neutrophils drug effects, Platelet Activating Factor analogs & derivatives, Platelet Activating Factor pharmacology, Platelet Activation drug effects
- Abstract
Platelet-activating factor (PAF) is a potent, bioactive phospholipid that acts on multiple cells and tissues through its G protein-coupled receptor (GPCR). PAF is not stored but is rapidly generated via enzymatic acetylation of the precursor 1-O-hexadecyl-2-hydroxy-sn-glycero-3-phosphocholine (lysoPAF). The bioactivity of PAF is effectively and tightly regulated by PAF acetylhydrolases, which convert PAF back to lysoPAF. Previous studies report that lysoPAF is an inactive precursor and metabolite of PAF. However, lysoPAF has not been carefully studied in its own context. Here we report that lysoPAF has an opposing effect of PAF in the activation of neutrophils and platelets. Whereas PAF potentiates neutrophil NADPH oxidase activation, lysoPAF dose-dependently inhibits this function. Inhibition by lysoPAF is not affected by the use of a PAF receptor antagonist or genetic deletion of the PAF receptor gene. The mechanism of lysoPAF-mediated inhibition of neutrophils involves an elevation in the intracellular cAMP level, and pharmacological blockade of adenylyl cyclase completely reverses the inhibitory effect of lysoPAF. In addition, lysoPAF increases intracellular cAMP levels in platelets and inhibits thrombin-induced platelet aggregation, which can be reversed by inhibition of protein kinase A. These findings identify lysoPAF as a bioactive lipid with opposing functions of PAF and suggest a novel and intrinsic regulatory mechanism for balance of the potent activity of PAF.
- Published
- 2009
- Full Text
- View/download PDF
143. PML/RARalpha fusion protein mediates the unique sensitivity to arsenic cytotoxicity in acute promyelocytic leukemia cells: Mechanisms involve the impairment of cAMP signaling and the aberrant regulation of NADPH oxidase.
- Author
-
Li L, Wang J, Ye RD, Shi G, Jin H, Tang X, and Yi J
- Subjects
- Arsenic Trioxide, Cell Survival drug effects, Dose-Response Relationship, Drug, Humans, Leukemia, Promyelocytic, Acute pathology, Oncogene Proteins, Fusion genetics, Reactive Oxygen Species metabolism, Time Factors, Transfection, U937 Cells, Antineoplastic Agents pharmacology, Apoptosis drug effects, Arsenicals pharmacology, Cyclic AMP metabolism, Leukemia, Promyelocytic, Acute enzymology, NADPH Oxidases metabolism, Oncogene Proteins, Fusion metabolism, Oxides pharmacology, Signal Transduction drug effects
- Abstract
Acute promyelocytic leukemia (APL) cells are characterized by PML/RARalpha fusion protein, high responsiveness to arsenic trioxide (ATO)-induced cytotoxicity and an abundant generation of reactive oxygen species (ROS). In this study we investigated the association among these three features in APL-derived NB4 cells. We found that NADPH oxidase-derived ROS generation was more abundant in NB4 cells compared with monocytic leukemia U937 cells. By using PR9, a sub-line of U937 stably transduced with the inducible PML/RARalpha expression vectors, we attributed disparities on ROS generation and ATO sensitivity to the occurrence of PML/RARalpha fusion protein, since PML/RARalpha-expressing cells appeared higher NADPH oxidase activity, higher ROS level and higher sensitivity to ATO. On the other hand, the basal intensity of cAMP signaling pathway was compared between NB4 and U937 as well as between PR9 cells with or without PML/RARalpha, demonstrating that PML/RARalpha-expressing cells had an impaired cAMP signaling pathway which relieved its inhibitory effect on NADPH oxidase derived ROS generation. In summary, the present study demonstrated the correlation of PML/RARalpha with cAMP signaling pathway, NADPH oxidase and ROS generation in APL cells. PML/RARalpha that bestows NB4 cells various pathological features, paradoxically also endows these cells with the basis for susceptibility to ATO-induced cytotoxcity., ((c) 2008 Wiley-Liss, Inc)
- Published
- 2008
- Full Text
- View/download PDF
144. Identification of novel formyl peptide receptor-like 1 agonists that induce macrophage tumor necrosis factor alpha production.
- Author
-
Schepetkin IA, Kirpotina LN, Tian J, Khlebnikov AI, Ye RD, and Quinn MT
- Subjects
- Animals, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Line, Tumor, Cells, Cultured, Dose-Response Relationship, Drug, Humans, Macrophages drug effects, Macrophages, Peritoneal drug effects, Macrophages, Peritoneal metabolism, Mice, Mice, Inbred BALB C, Pharmaceutical Preparations administration & dosage, Pharmaceutical Preparations chemistry, Rats, Receptors, Formyl Peptide biosynthesis, Receptors, Lipoxin biosynthesis, Macrophages metabolism, Receptors, Formyl Peptide agonists, Receptors, Lipoxin agonists, Tumor Necrosis Factor-alpha biosynthesis
- Abstract
Development of immunomodulatory agents that enhance innate immune responses represents a promising strategy for combating infectious diseases. In the present studies, we screened a series of 71 arylcarboxylic acid hydrazide derivatives for their ability to induce macrophage tumor necrosis factor alpha (TNF-alpha) production and identified six such compounds, including one compound previously shown to be a formyl peptide receptor (FPR/FPRL1) agonist. The two most potent compounds [compound 1, nicotinic acid [5-(3-bromophenyl)-2-furyl]methylene-hydrazide; compound 2, 4-fluoro-benzoic acid [5-(3-trifluoromethyl-phenyl)-2-furyl]-methylene-hydrazide] were selected for further analysis. These compounds induced de novo production of TNF-alpha in a dose- and time-dependent manner in human and murine monocyte/macrophage cell lines and in primary macrophages. These compounds also induced mobilization of intracellular Ca(2+), production of reactive oxygen species, and chemotaxis in human and murine phagocytes. Induction of macrophage TNF-alpha production was pertussis toxin-sensitive, and analysis of the cellular target of these compounds showed that they were FPRL1-specific agonists and that this response was blocked by FPR/FPRL1 and FPRL1-specific antagonists. In addition, pharmacophore modeling showed a high degree of similarity for low-energy conformations of these two compounds to the current pharmacophore model for FPR ligands ( Mol Pharmacol 68: 1301-1310, 2005 ). Overall, these compounds represent novel FPRL1 agonists that induce TNF-alpha, a response distinct from those induced by other known FPR and FPRL1 agonists.
- Published
- 2008
- Full Text
- View/download PDF
145. Identification of formyl peptides from Listeria monocytogenes and Staphylococcus aureus as potent chemoattractants for mouse neutrophils.
- Author
-
Southgate EL, He RL, Gao JL, Murphy PM, Nanamori M, and Ye RD
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Mice, Knockout, N-Formylmethionine Leucyl-Phenylalanine immunology, Neutrophil Activation, Neutrophils metabolism, Oligopeptides metabolism, Rats, Receptors, Formyl Peptide deficiency, Receptors, Formyl Peptide immunology, Transfection, Chemotactic Factors immunology, Chemotaxis, Leukocyte, Listeria monocytogenes immunology, Neutrophils immunology, Oligopeptides immunology, Receptors, Formyl Peptide metabolism, Staphylococcus aureus immunology
- Abstract
The prototypic formyl peptide N-formyl-Met-Leu-Phe (fMLF) is a major chemoattractant found in Escherichia coli culture supernatants and a potent agonist at human formyl peptide receptor (FPR) 1. Consistent with this, fMLF induces bactericidal functions in human neutrophils at nanomolar concentrations. However, it is a much less potent agonist for mouse FPR (mFPR) 1 and mouse neutrophils, requiring micromolar concentrations for cell activation. To determine whether other bacteria produce more potent agonists for mFPR1, we examined formyl peptides from Listeria monocytogenes and Staphylococcus aureus for their abilities to activate mouse neutrophils. A pentapeptide (N-formyl-Met-Ile-Val-Ile-Leu (fMIVIL)) from L. monocytogenes and a tetrapeptide (N-formyl-Met-Ile-Phe-Leu (fMIFL)) from S. aureus were found to induce mouse neutrophil chemotaxis at 1-10 nM and superoxide production at 10-100 nM, similar to the potency of fMLF on human neutrophils. Using transfected cell lines expressing mFPR1 and mFPR2, which are major forms of FPRs in mouse neutrophils, we found that mFPR1 is responsible for the high potency of fMIVIL and fMIFL. In comparison, activation of mFPR2 requires micromolar concentrations of the two peptides. Genetic deletion of mfpr1 resulted in abrogation of neutrophil superoxide production and degranulation in response to fMIVIL and fMIFL, further demonstrating that mFPR1 is the primary receptor for detection of these formyl peptides. In conclusion, the formyl peptides from L. monocytogenes and S. aureus are approximately 100-fold more potent than fMLF in activating mouse neutrophils. The ability of mFPR1 to detect bacterially derived formyl peptides indicates that this important host defense mechanism is conserved in mice.
- Published
- 2008
- Full Text
- View/download PDF
146. Cutting edge: TLR2 is a functional receptor for acute-phase serum amyloid A.
- Author
-
Cheng N, He R, Tian J, Ye PP, and Ye RD
- Subjects
- Animals, Cell Line, Cricetinae, Cytokines biosynthesis, Cytokines genetics, Humans, Mice, Protein Binding, Signal Transduction, Toll-Like Receptor 2 deficiency, Toll-Like Receptor 2 genetics, Serum Amyloid A Protein metabolism, Toll-Like Receptor 2 metabolism
- Abstract
Induced secretion of acute-phase serum amyloid A (SAA) is a host response to danger signals and a clinical indication of inflammation. The biological functions of SAA in inflammation have not been fully defined, although recent reports indicate that SAA induces proinflammatory cytokine expression. We now show that TLR2 is a functional receptor for SAA. HeLa cells expressing TLR2 responded to SAA with potent activation of NF-kappaB, which was enhanced by TLR1 expression and blocked by the Toll/IL-1 receptor/resistance (TIR) deletion mutants of TLR1, TLR2, and TLR6. SAA stimulation led to increased phosphorylation of MAPKs and accelerated IkappaBalpha degradation in TLR2-HeLa cells, and results from a solid-phase binding assay showed SAA interaction with the ectodomain of TLR2. Selective reduction of SAA-induced gene expression was observed in tlr2-/- mouse macrophages compared with wild-type cells. These results suggest a potential role for SAA in inflammatory diseases through activation of TLR2.
- Published
- 2008
- Full Text
- View/download PDF
147. Neutrophil caveolin-1 expression contributes to mechanism of lung inflammation and injury.
- Author
-
Hu G, Ye RD, Dinauer MC, Malik AB, and Minshall RD
- Subjects
- Animals, Cell Adhesion drug effects, Cell Line, Chemotaxis drug effects, Endothelial Cells cytology, Endothelial Cells drug effects, Enzyme Activation drug effects, Gene Deletion, In Vitro Techniques, Lung blood supply, Lung drug effects, Lung metabolism, Lung pathology, Mice, N-Formylmethionine Leucyl-Phenylalanine pharmacology, NADPH Oxidases metabolism, Neutrophils cytology, Neutrophils drug effects, Neutrophils enzymology, Permeability drug effects, Pulmonary Edema pathology, Superoxides metabolism, rac GTP-Binding Proteins metabolism, rac1 GTP-Binding Protein metabolism, RAC2 GTP-Binding Protein, Caveolin 1 metabolism, Neutrophils metabolism, Pneumonia metabolism, Pneumonia pathology
- Abstract
Caveolin-1 present in immune cells may be involved in regulation of the inflammatory response. Here, using caveolin-1-null (Cav-1(-/-)) mice, we addressed the role of caveolin-1 in polymorphonuclear neutrophils (PMNs) in regulating PMN activation-mediated lung injury. In lungs of wild-type (Cav-1(+/+)) mice perfused at constant flow with Krebs-Henseleit solution, addition of Cav-1(+/+) PMNs (4 x 10(6) cells) into the perfusate followed by their activation with formyl-Met-Leu-Phe (fMLP, 1.0 muM) plus platelet-activating factor (1.0 nM) increased pulmonary microvessel filtration coefficient by 150% and wet-to-dry lung weight ratio by 50% as well as PMN accumulation in lungs. These responses were markedly reduced in lungs perfused with Cav-1(-/-) PMNs followed by addition of the same activating agents. fMLP-stimulated adhesion of Cav-1(-/-) PMNs to pulmonary microvascular endothelial cells and migration of Cav-1(-/-) PMNs across endothelial monolayers were also impaired compared with Cav-1(+/+) PMNs. Cav-1(-/-) PMNs showed 50-80% reduction in PMA- or fMLP-stimulated superoxide production compared with Cav-1(+/+) PMNs. In addition, Cav-1(-/-) PMNs had decreased migratory activity (50%) and adhesion to fibrinogen (40%) in response to fMLP. Rac1 and Rac2 were activated in Cav-1(+/+) PMNs after stimulation of fMLP but not in Cav-1(-/-) PMNs. Exogenous expression of caveolin-1 in COS-phox cells augmented the fMLP-induced Rac1 activation and superoxide production, indicating a direct role of caveolin-1 in the mechanism of superoxide production. Thus caveolin-1 expression in PMNs plays a key role in mediating PMN activation, adhesion, and transendothelial migration and in PMN activation-induced lung inflammation and vascular injury.
- Published
- 2008
- Full Text
- View/download PDF
148. A critical role of protein kinase C delta activation loop phosphorylation in formyl-methionyl-leucyl-phenylalanine-induced phosphorylation of p47(phox) and rapid activation of nicotinamide adenine dinucleotide phosphate oxidase.
- Author
-
Cheng N, He R, Tian J, Dinauer MC, and Ye RD
- Subjects
- Animals, COS Cells, Catalysis, Cell Line, Chlorocebus aethiops, Humans, Mice, Phosphorylation drug effects, Protein Kinase C-delta immunology, Substrate Specificity, Superoxides metabolism, N-Formylmethionine Leucyl-Phenylalanine pharmacology, NADPH Oxidases metabolism, Protein Kinase C-delta metabolism
- Abstract
Generation of superoxide by professional phagocytes is an important mechanism of host defense against bacterial infection. Several protein kinase C (PKC) isoforms have been found to phosphorylate p47(phox), resulting in its membrane translocation and activation of the NADPH oxidase. However, the mechanism by which specific PKC isoforms regulate NADPH oxidase activation remains to be elucidated. In this study, we report that PKCdelta phosphorylation in its activation loop is rapidly induced by fMLF and is essential for its ability to catalyze p47(phox) phosphorylation. Using transfected COS-7 cells expressing gp91(phox), p22(phox), p67(phox), and p47(phox) (COS-phox cells), we found that a functionally active PKCdelta is required for p47(phox) phosphorylation and reconstitution of NADPH oxidase. PKCbetaII cannot replace PKCdelta for this function. Characterization of PKCdelta/PKCbetaII chimeras has led to the identification of the catalytic domain of PKCdelta as a target of regulation by fMLF, which induces a biphasic (30 and 180 s) phosphorylation of Thr(505) in the activation loop of mouse PKCdelta. Mutation of Thr(505) to alanine abolishes the ability of PKCdelta to catalyze p47(phox) phosphorylation in vitro and to reconstitute NADPH oxidase in the transfected COS-phox cells. A correlation between fMLF-induced activation loop phosphorylation and superoxide production is also established in the differentiated PLB-985 human myelomonoblastic cells. We conclude that agonist-induced PKCdelta phosphorylation is a novel mechanism for NADPH oxidase activation. The ability to induce PKCdelta phosphorylation may distinguish a full agonist from a partial agonist for superoxide production.
- Published
- 2007
- Full Text
- View/download PDF
149. Characterization of a mutation in the Phox homology domain of the NADPH oxidase component p40phox identifies a mechanism for negative regulation of superoxide production.
- Author
-
Chen J, He R, Minshall RD, Dinauer MC, and Ye RD
- Subjects
- Actins chemistry, Animals, Biological Transport, COS Cells, Cell Line, Tumor, Cell Membrane metabolism, Chlorocebus aethiops, Humans, Microfilament Proteins chemistry, Protein Structure, Tertiary, Superoxides chemistry, Superoxides metabolism, Gene Expression Regulation, Homeodomain Proteins genetics, Homeodomain Proteins physiology, Mutation, NADPH Oxidases metabolism, Phosphoproteins chemistry
- Abstract
The phagocyte oxidase (Phox) protein p40(phox) contains a Phox homology (PX) domain which, when expressed alone, interacts with phosphatidylinositol 3-phosphate (PtdIns (3)P). The functions of the PX domain in p40(phox) localization, association with the cytoskeleton, and superoxide production were examined in transgenic COS-7 cells expressing gp91(phox), p22(phox), p67(phox), and p47(phox) (COS(phox) cells). Full-length p40(phox) exhibited a cytoplasmic localization pattern in resting cells. Upon stimulation with phorbol 12-myristate 13-acetate or fMet-Leu-Phe, p40(phox) translocated to plasma membrane in a p67(phox)- and p47(phox)-dependent manner. Heterologous expression of p40(phox) markedly enhanced superoxide production in phorbol 12-myristate 13-acetate - and fMet-Leu-Phe-stimulated COS(phox) cells. Unexpectedly, mutation of Arg-57 in the PX domain to Gln, which abrogated PtdIns (3)P binding, produced a dominant inhibitory effect on agonist-induced superoxide production and membrane translocation of p47(phox) and p67(phox). The mutant p40(phox) (p40R57Q) displayed increased association with actin and moesin and was found enriched in the Triton X-100-insoluble fraction along with p67(phox) and p47(phox). The enhanced cytoskeleton association of p67(phox) and p47(phox) and the dominant inhibitory effect produced by the p40R57Q were alleviated when a second mutation at Asp-289, which eliminated p40(phox) interaction with p67(phox), was introduced. Likewise, cytochalasin B treatment abolished the dominant inhibitory effect of p40R57Q on superoxide production. These findings suggest a dual regulatory mechanism through the PX domain of p40(phox); its interaction with the actin cytoskeleton may stabilize NADPH oxidase in resting cells, and its binding of PtdIns (3)P potentiates superoxide production upon agonist stimulation. Both functions require the association of p40(phox) with p67(phox).
- Published
- 2007
- Full Text
- View/download PDF
150. Pharmacological characterization of a novel nonpeptide antagonist for formyl peptide receptor-like 1.
- Author
-
Zhou C, Zhang S, Nanamori M, Zhang Y, Liu Q, Li N, Sun M, Tian J, Ye PP, Cheng N, Ye RD, and Wang MW
- Subjects
- Arachidonic Acid pharmacology, Calcium metabolism, Cell Line, Chemotaxis drug effects, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Humans, Ligands, Phosphorylation, Receptors, Formyl Peptide genetics, Chemotactic Factors pharmacology, Oligopeptides pharmacology, Receptors, Formyl Peptide antagonists & inhibitors
- Abstract
A series of quinazolinone derivatives were synthesized based on a hit compound identified from a high-throughput screening campaign targeting the human formyl peptide receptor-like 1 (FPRL1). Based on structure-activity relationship analysis, we found that substitution on the para position of the 2-phenyl group of the quinazolinone backbone could alter the pharmacological properties of the compound. The methoxyl substitution produced an agonist 4-butoxy-N-[2-(4-methoxy-phenyl)-4-oxo-1,4-dihydro-2H-quinazolin-3-yl]-benzamide (Quin-C1; C1), whereas a hydroxyl substitution resulted in a pure antagonist, Quin-C7 (C7). Several partial agonists were derived from other substitutions on the para position. C7 partially displaced [(125)I]Trp-Lys-Tyr-Met-Val-d-Met-NH(2) (WKYMVm) binding to FPRL1 but not [(3)H]N-formyl-Met-Leu-Phe to formyl peptide receptor. In functional assays using FPRL1-expressing RBL-2H3 cells, C7 inhibited calcium mobilization and chemotaxis induced by WKYMVm and C1 and degranulation elicited by C1. C7 also suppressed C1-induced extracellular signal-regulated kinase phosphorylation and reduced arachidonic acid-induced ear edema in mice. This study represents the first characterization of a nonpeptidic antagonist for FPRL1 and suggests the prospect of using low molecular weight compounds as modulators of chemoattractant receptors in vitro and in vivo.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.