Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Gen Physiol ; 156(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38771271

ABSTRACT

The voltage-sensing domain (VSD) is a four-helix modular protein domain that converts electrical signals into conformational changes, leading to open pores and active enzymes. In most voltage-sensing proteins, the VSDs do not interact with one another, and the S1-S3 helices are considered mainly scaffolding, except in the voltage-sensing phosphatase (VSP) and the proton channel (Hv). To investigate its contribution to VSP function, we mutated four hydrophobic amino acids in S1 to alanine (F127, I131, I134, and L137), individually or in combination. Most of these mutations shifted the voltage dependence of activity to higher voltages; however, not all substrate reactions were the same. The kinetics of enzymatic activity were also altered, with some mutations significantly slowing down dephosphorylation. The voltage dependence of VSD motions was consistently shifted to lower voltages and indicated a second voltage-dependent motion. Additionally, none of the mutations broke the VSP dimer, indicating that the S1 impact could stem from intra- and/or intersubunit interactions. Lastly, when the same mutations were introduced into a genetically encoded voltage indicator, they dramatically altered the optical readings, making some of the kinetics faster and shifting the voltage dependence. These results indicate that the S1 helix in VSP plays a critical role in tuning the enzyme's conformational response to membrane potential transients and influencing the function of the VSD.


Subject(s)
Phosphoric Monoester Hydrolases , Animals , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/chemistry , Hydrophobic and Hydrophilic Interactions , Mutation , Protein Domains , Kinetics , Humans , Phosphorylation
2.
bioRxiv ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38234747

ABSTRACT

The voltage sensing domain (VSD) is a four-helix modular protein domain that converts electrical signals into conformational changes, leading to open pores and active enzymes. In most voltage sensing proteins, the VSDs do not interact with one another and the S1-S3 helices are considered mainly as scaffolding. The two exceptions are the voltage sensing phosphatase (VSP) and the proton channel (Hv). VSP is a voltage-regulated enzyme and Hvs are channels that only have VSDs. To investigate the S1 contribution to VSP function, we individually mutated four hydrophobic amino acids in S1 to alanine (F127, I131, I134 and L137). We also combined these mutations to generate quadruple mutation designated S1-Q. Most of these mutations shifted the voltage dependence of activity to higher voltages though interestingly, not all substrate reactions were the same. The kinetics of enzymatic activity were also altered with some mutations significantly slowing down dephosphorylation. The voltage dependence of VSD motions were consistently shifted to lower voltages and indicated a second voltage dependent motion. Co-immunoprecipitation demonstrated that none of the mutations broke the VSP dimer indicating that the S1 impact could stem from intrasubunit and/or intersubunit interactions. Lastly, when the same alanine mutations were introduced into a genetically encoded voltage indicator, they dramatically altered the optical readings, making some of the kinetics faster and shifting the voltage dependence. These results indicate that the S1 helix in VSP plays a critical role in tuning the enzymes conformational response to membrane potential transients and influencing the function of the VSD.

3.
Mol Cell ; 74(1): 132-142.e5, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30872121

ABSTRACT

Bacteria and archaea have evolved sophisticated adaptive immune systems that rely on CRISPR RNA (crRNA)-guided detection and nuclease-mediated elimination of invading nucleic acids. Here, we present the cryo-electron microscopy (cryo-EM) structure of the type I-F crRNA-guided surveillance complex (Csy complex) from Pseudomonas aeruginosa bound to a double-stranded DNA target. Comparison of this structure to previously determined structures of this complex reveals a ∼180-degree rotation of the C-terminal helical bundle on the "large" Cas8f subunit. We show that the double-stranded DNA (dsDNA)-induced conformational change in Cas8f exposes a Cas2/3 "nuclease recruitment helix" that is structurally homologous to a virally encoded anti-CRISPR protein (AcrIF3). Structural homology between Cas8f and AcrIF3 suggests that AcrIF3 is a mimic of the Cas8f nuclease recruitment helix.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA, Bacterial/metabolism , Molecular Mimicry , Pseudomonas aeruginosa/enzymology , RNA, Bacterial/metabolism , RNA, Guide, Kinetoplastida/metabolism , Viral Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/immunology , Cryoelectron Microscopy , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
4.
PLoS One ; 13(7): e0200444, 2018.
Article in English | MEDLINE | ID: mdl-29990379

ABSTRACT

Neutrophils are the most abundant white blood cells, with a vital role in innate immune defense against bacterial and fungal pathogens. Although mostly associated with pathological processes directly related to immune defense, they can also play a detrimental role in inflammatory conditions and have been found to have a pro-metastatic role in the spread of cancer cells. Here, we explore ways to temporarily suppress these detrimental activities. We first examined the possibility of using siRNA and antisense oligonucleotides (ASOs) for transient knockdown of the human and mouse C5a receptor, an important chemoattractant receptor involved in neutrophil-mediated injury that is associated with myocardial infarction, sepsis, and neurodegenerative diseases. We found that siRNAs and ASOs transfected into cultured cell lines can eliminate 70-90% of C5a receptor mRNA and protein within 72 h of administration, a clinically relevant time frame after a cardiovascular event. Targeted drug delivery to specific cells or tissues of interest in a mammalian host, however, remains a major challenge. Here, using phage display technology, we have identified peptides that bind specifically to CD177, a neutrophil-specific surface molecule. We have attached these peptides to fluorescent, lipid-based nanoparticles and confirmed targeting and delivery to cultured cells ectopically presenting either human or mouse CD177. In addition, we have shown peptide-nanoparticle binding specifically to neutrophils in human and mouse blood. We anticipate that these or related tagged nanoparticles may be therapeutically useful for delivery of siRNAs or ASOs to neutrophils for transient knockdown of pro-inflammatory proteins such as the C5a receptor.


Subject(s)
Isoantigens/metabolism , Nanoparticles/administration & dosage , Neutrophils/metabolism , Receptors, Cell Surface/metabolism , Animals , CHO Cells , Cricetulus , GPI-Linked Proteins/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Neutrophils/cytology , Oligonucleotides, Antisense/administration & dosage , Protein Binding , RNA, Small Interfering , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism
5.
ACS Nano ; 12(2): 942-953, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29131580

ABSTRACT

The assembly of individual molecules into hierarchical structures is a promising strategy for developing three-dimensional materials with properties arising from interaction between the individual building blocks. Virus capsids are elegant examples of biomolecular nanostructures, which are themselves hierarchically assembled from a limited number of protein subunits. Here, we demonstrate the bio-inspired modular construction of materials with two levels of hierarchy: the formation of catalytically active individual virus-like particles (VLPs) through directed self-assembly of capsid subunits with enzyme encapsulation, and the assembly of these VLP building blocks into three-dimensional arrays. The structure of the assembled arrays was successfully altered from an amorphous aggregate to an ordered structure, with a face-centered cubic lattice, by modifying the exterior surface of the VLP without changing its overall morphology, to modulate interparticle interactions. The assembly behavior and resultant lattice structure was a consequence of interparticle interaction between exterior surfaces of individual particles and thus independent of the enzyme cargos encapsulated within the VLPs. These superlattice materials, composed of two populations of enzyme-packaged VLP modules, retained the coupled catalytic activity in a two-step reaction for isobutanol synthesis. This study demonstrates a significant step toward the bottom-up fabrication of functional superlattice materials using a self-assembly process across multiple length scales and exhibits properties and function that arise from the interaction between individual building blocks.


Subject(s)
Alcohol Dehydrogenase/metabolism , Carboxy-Lyases/metabolism , Alcohol Dehydrogenase/chemistry , Biocatalysis , Carboxy-Lyases/chemistry , Particle Size , Surface Properties
6.
Nanoscale ; 9(29): 10420-10430, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28702648

ABSTRACT

Virus-like particles (VLPs) derived from the bacteriophage P22 offer an interesting and malleable platform for encapsulation and multivalent presentation of cargo molecules. The packaging of cargo in P22 VLP is typically achieved through genetically enabled directed in vivo encapsulation. However, this approach does not allow control over the packing density and composition of the encapsulated cargos. Here, we have adopted an in vitro assembly approach to gain control over cargo packaging in P22. The packaging was controlled by closely regulating the stoichiometric ratio of cargo-fused-scaffold protein and wild-type scaffold protein during the in vitro assembly. In a "one-pot assembly reaction" coat protein subunits were incubated with varied ratios of wild-type scaffold protein and cargo-fused-scaffold protein, which resulted in the encapsulation of both components in a co-assembled capsid. These experiments demonstrate that an input stoichiometry can be used to achieve controlled packaging of multiple cargos within the VLP. The porous nature of P22 allows the escape and re-entry of wild-type scaffold protein from the assembled capsid but scaffold protein fused to a protein-cargo cannot traverse the capsid shell due to the size of the cargo. This has allowed us to control and alter the packing density by selectively releasing wild-type scaffold protein from the co-assembled capsids. We have demonstrated these concepts in the P22 system using an encapsulated streptavidin protein and have shown its highly selective interaction with biotin or biotin derivatives. Additionally, this system can be used to encapsulate small molecules coupled to biotin, or display large proteins, that cannot enter the capsid and thus remain available for the multivalent display on the exterior of the capsid when attached to a flexible biotinylated linker. Thus, we have developed a P22 system with controlled protein cargo composition and packing density, to which both small and large molecules can be attached at high copy number on the interior or exterior of the capsid.


Subject(s)
Bacteriophage P22 , Capsid Proteins/chemistry , Capsid/chemistry , Drug Carriers/chemistry
7.
Mol Pharm ; 13(3): 1191-6, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26894836

ABSTRACT

Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) RNA-guided endonucleases are powerful new tools for targeted genome engineering. These nucleases provide an efficient and precise method for manipulating eukaryotic genomes; however, delivery of these reagents to specific cell-types remains challenging. Virus-like particles (VLPs) derived from bacteriophage P22, are robust supramolecular protein cage structures with demonstrated utility for cell type-specific delivery of encapsulated cargos. Here, we genetically fuse Cas9 to a truncated form of the P22 scaffold protein, which acts as a template for capsid assembly as well as a specific encapsulation signal for Cas9. Our results indicate that Cas9 and a single-guide RNA are packaged inside the P22 VLP, and activity assays indicate that this RNA-guided endonuclease is functional for sequence-specific cleavage of dsDNA targets. This work demonstrates the potential for developing P22 as a delivery vehicle for cell specific targeting of Cas9.


Subject(s)
Bacterial Proteins/metabolism , Bacteriophage P22/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Drug Carriers/chemistry , Drug Delivery Systems , Endonucleases/metabolism , Genetic Engineering/methods , Nanoparticles/chemistry , CRISPR-Associated Protein 9 , Endonucleases/genetics , Humans , RNA Editing/genetics
8.
Nat Chem ; 8(2): 179-85, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26791902

ABSTRACT

The chemistry of highly evolved protein-based compartments has inspired the design of new catalytically active materials that self-assemble from biological components. A frontier of this biodesign is the potential to contribute new catalytic systems for the production of sustainable fuels, such as hydrogen. Here, we show the encapsulation and protection of an active hydrogen-producing and oxygen-tolerant [NiFe]-hydrogenase, sequestered within the capsid of the bacteriophage P22 through directed self-assembly. We co-opted Escherichia coli for biomolecular synthesis and assembly of this nanomaterial by expressing and maturing the EcHyd-1 hydrogenase prior to expression of the P22 coat protein, which subsequently self assembles. By probing the infrared spectroscopic signatures and catalytic activity of the engineered material, we demonstrate that the capsid provides stability and protection to the hydrogenase cargo. These results illustrate how combining biological function with directed supramolecular self-assembly can be used to create new materials for sustainable catalysis.


Subject(s)
Escherichia coli/chemistry , Hydrogen/chemistry , Hydrogenase/chemistry , Catalysis
9.
ACS Biomater Sci Eng ; 2(12): 2324-2332, 2016.
Article in English | MEDLINE | ID: mdl-29367948

ABSTRACT

Subunit vaccines provide a safe, focused alternative to conventional vaccines. However, these vaccines often require significant adjuvants and are particularly hard to target toward cytotoxic T lymphocyte (CTL) immunity. Viruslike particles (VLPs) provide biomaterial scaffolds with pathogen-like polyvalent structures making them useful platforms for biomimetic antigen delivery to the immune system. Encapsidation of antigens within VLPs has been shown to enhance antigen availability for CD8 T cell responses. Here, we examine the potential to generate complex responses to multiple subunit antigens localized within the same VLP particle. Two proteins of respiratory syncytial virus (RSV) with well-characterized CD8 T cell responses, the matrix (M) and matrix 2 (M2) proteins, were successfully coencapsidated within the P22 VLP. Upon intranasal administration in mice, the particles stimulated CD8 T cell memory responses against both antigens. In addition, vaccination elicited tissue-resident T cell populations. Upon subsequent RSV challenge, P22-M/M2-treated mice displayed significantly reduced lung viral titers. This demonstrates the utility of the P22 VLP in directing immune responses to multiple encapsidated viral antigens, demonstrating the potential of this technology to facilitate immunity to multiple targets simultaneously.

10.
ACS Nano ; 9(9): 9134-47, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26266824

ABSTRACT

Viruses use spatial control of constituent proteins as a means of manipulating and evading host immune systems. Similarly, precise spatial control of proteins encapsulated or presented on designed nanoparticles has the potential to biomimetically amplify or shield biological interactions. Previously, we have shown the ability to encapsulate a wide range of guest proteins within the virus-like particle (VLP) from Salmonella typhimurium bacteriophage P22, including antigenic proteins from human pathogens such as influenza. Expanding on this robust encapsulation strategy, we have used the trimeric decoration protein (Dec) from bacteriophage L as a means of controlled exterior presentation on the mature P22 VLP, to which it binds with high affinity. Through genetic fusion to the C-terminus of the Dec protein, either the 17 kDa soluble region of murine CD40L or a minimal peptide designed from the binding region of the "self-marker" CD47 was independently presented on the P22 VLP capsid exterior. Both candidates retained function when presented as a Dec-fusion. Binding of the Dec domain to the P22 capsid was minimally changed across designed constructs, as measured by surface plasmon resonance, demonstrating the broad utility of this presentation strategy. Dec-mediated presentation offers a robust, modular means of decorating the exposed exterior of the P22 capsid in order to further orchestrate responses to internally functionalized VLPs within biological systems.


Subject(s)
Bacteriophage P22/chemistry , Capsid Proteins/chemistry , Recombinant Fusion Proteins/genetics , Virion/chemistry , Animals , Bacteriophage P22/genetics , Bacteriophage lambda/chemistry , Bacteriophage lambda/genetics , CD40 Ligand/chemistry , CD40 Ligand/genetics , CD47 Antigen/chemistry , CD47 Antigen/genetics , Capsid Proteins/genetics , Humans , Mice , Peptides/chemistry , Peptides/genetics , Recombinant Fusion Proteins/chemistry , Salmonella typhimurium/virology , Virion/genetics
11.
PLoS One ; 6(12): e28712, 2011.
Article in English | MEDLINE | ID: mdl-22174875

ABSTRACT

The gene encoding the human formyl peptide receptor 1 (FPR1) is heterogeneous, containing numerous single nucleotide polymorphisms (SNPs). Here, we examine the effect of these SNPs on gene transcription and protein translation. We also identify gene promoter sequences and putative FPR1 transcription factors. To test the effect of codon bias and codon pair bias on FPR1 expression, four FPR1 genetic variants were expressed in human myeloid U937 cells fused to a reporter gene encoding firefly luciferase. No significant differences in luciferase activity were detected, suggesting that the translational regulation and protein stability of FPR1 are modulated by factors other than the SNP codon bias and the variant amino acid properties. Deletion and mutagenesis analysis of the FPR1 promoter showed that a CCAAT box is not required for gene transcription. A -88/41 promoter construct resulted in the strongest transcriptional activity, whereas a -72/41 construct showed large reduction in activity. The region between -88 and -72 contains a consensus binding site for the transcription factor PU.1. Mutagenesis of this site caused significant reduction in reporter gene expression. The PU.1 binding was confirmed in vivo by chromatin immunoprecipitation, and the binding to nucleotides -84 to -76 (TTCCTATTT) was confirmed in vitro by an electrophoretic mobility shift assay. Thus, similar to many other myeloid genes, FPR1 promoter activity requires PU.1. Two single nucleotide polymorphisms at -56 and -54 did not significantly affect FPR1 gene expression, despite differences in binding of transcription factor IRF1 in vitro. Inflammatory mediators such as interferon-γ, tumor necrosis factor-α, and lipopolysaccharide did not increase FPR1 promoter activity in myeloid cells, whereas differentiation induced by DMSO and retinoic acid enhanced the activity. This implies that the expression of FPR1 in myeloid cells is developmentally regulated, and that the differentiated cells are equipped for immediate response to microbial infections.


Subject(s)
Inflammation Mediators/metabolism , Polymorphism, Single Nucleotide/genetics , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/genetics , Transcription Factors/metabolism , Base Sequence , Binding Sites , Cell Differentiation/drug effects , Chromatin Immunoprecipitation , Codon/genetics , Dimethyl Sulfoxide/pharmacology , Gene Expression Regulation/drug effects , Genotype , Humans , Molecular Sequence Data , Mutagenesis/drug effects , Mutagenesis/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Reproducibility of Results , Transcription, Genetic/drug effects , Tretinoin/pharmacology , U937 Cells
12.
Biochim Biophys Acta ; 1793(2): 406-17, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18952127

ABSTRACT

Formyl peptide receptor (FPR) is a chemoattractant G protein-coupled receptor (GPCR) involved in the innate immune response against bacteria. Receptor activation is terminated by receptor phosphorylation of two serine- and threonine-rich regions located in the distal half of the cytoplasmic tail. In this study we show that introduction of an amino acid with a bulky side chain (leucine or glutamine) adjacent to a single leucine, L320, in the membrane-proximal half of the cytoplasmic tail, significantly enhanced receptor phosphorylation, beta-arrestin1/2 translocation, and receptor endocytosis, without affecting G(i)-mediated ERK1/2 activation and release of intracellular calcium. In addition, the point mutations resulted in diminished susceptibility to trypsin, suggesting a conformation different from that of wild type FPR. Alignment of the FPR sequence with the rhodopsin sequence showed that L320 resides immediately C-terminal of an amphipathic region that in rhodopsin forms helix 8. Deletion of seven amino acids (Delta309-315) from the predicted helix 8 of FPR (G307-S319) caused reduced cell signaling as well as defects in receptor phosphorylation, beta-arrestin1/2 translocation and endocytosis. Thus, the amino acid content in the N-terminal half of the cytoplasmic tail influences the structure and desensitization of FPR.


Subject(s)
Cell Membrane/metabolism , Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/metabolism , Amino Acid Sequence , Animals , Arrestins/metabolism , CHO Cells , Cricetinae , Cricetulus , Endocytosis , GTP-Binding Proteins/metabolism , Humans , Leucine/metabolism , Ligands , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation , Protein Binding , Protein Transport , Receptors, Formyl Peptide/agonists , Signal Transduction , Structure-Activity Relationship , Trypsin/metabolism , beta-Arrestins
13.
Biochim Biophys Acta ; 1783(6): 1261-70, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18346468

ABSTRACT

The carboxyl tail of G protein-coupled receptors contains motifs that regulate receptor interactions with intracellular partners. Activation of the human neutrophil complement fragment C5a receptor (C5aR) is terminated by phosphorylation of the carboxyl tail followed by receptor internalization. In this study, we demonstrated that bulky hydrophobic residues in the membrane-proximal region of the C5aR carboxyl tail play an important role in proper structure and function of the receptor: Substitution of leucine 319 with alanine (L319A) resulted in receptor retention in the endoplasmic reticulum, whereas a L318A substitution allowed receptor transport to the cell surface, but showed slow internalization upon activation, presumably due to a defect in phosphorylation by both PKC and GRK. Normal agonist-induced activation of ERK1/2 and intracellular calcium release suggested that the L318A mutation did not affect receptor signaling. Binding of GRK2 and PKCbetaII to intracellular loop 3 of C5aR in vitro indicated that mutagenesis of L318 did not affect kinase binding. Limited proteolysis with trypsin revealed a conformational difference between wild type and mutant receptor. Our studies support a model in which the L318/L319 stabilizes an amphipathic helix (Q305-R320) in the membrane-proximal region of C5aR.


Subject(s)
Endocytosis , Leucine/chemistry , Receptors, Complement/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Blotting, Western , CHO Cells , Calcium/metabolism , Cell Membrane/metabolism , Cricetinae , Cricetulus , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique, Indirect , G-Protein-Coupled Receptor Kinase 2/metabolism , Humans , Leucine/genetics , Leucine/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Neutrophils/metabolism , Phosphorylation , Protein Conformation , Protein Kinase C/metabolism , Protein Kinase C beta , Radioligand Assay , Receptor, Anaphylatoxin C5a , Receptors, Complement/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
14.
Immunogenetics ; 60(2): 83-93, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18253729

ABSTRACT

The chemoattractant neutrophil formyl peptide receptor (FPR) binds bacterial and mitochondrial N-formylated peptides, which allows the neutrophils to find the bacterial source and/or site of tissue damage. Certain inflammatory disorders may be due in part to an impaired innate immune system that does not respond to acute bacterial damage in a timely fashion. Because the human FPR is encoded by a large number of different haplotypes arising from ten single-nucleotide polymorphisms, we examined the possibility that some of these haplotypes are functionally distinct. We analyzed the response of three common FPR haplotypes to peptides from Escherichia coli, Mycobacterium avium ssp. paratuberculosis, and human mitochondria. All three haplotypes responded similarly to the E. coli and mitochondrial peptides, whereas one required a higher concentration of the M. avium peptide fMFEDAVAWF for receptor downregulation, receptor signaling, and chemotaxis. This raises the possibility of additional bacterial species differences in functional responses among FPR variants and establishes a precedent with potentially important implications for our innate immune response against bacterial infections. We also investigated whether certain FPR haplotypes are associated with rheumatoid arthritis (RA) by sequencing FPR1 from 148 Caucasian individuals. The results suggested that FPR haplotypes do not significantly contribute toward RA.


Subject(s)
Arthritis, Rheumatoid/genetics , Haplotypes/genetics , Peptide Fragments/pharmacology , Receptors, Formyl Peptide/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , CHO Cells , Chemotactic Factors , Cricetinae , Cricetulus , Escherichia coli/metabolism , Female , Fluorescent Antibody Technique , Genotype , Humans , Middle Aged , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Mycobacterium avium/metabolism , Paratuberculosis/metabolism , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Receptors, Formyl Peptide/immunology , Transfection
15.
Cell Signal ; 20(2): 424-31, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18060741

ABSTRACT

Formyl peptide receptor (FPR) and C5a receptor (C5aR) are chemoattractant G protein-coupled receptors (GPCRs) involved in the innate immune response against bacterial infections and tissue injury. Like other GPCRs, they recruit beta-arrestin1/2 to the plasma membrane and activate the extracellular signal-regulated kinases 1 and 2 (ERK1/2). Previous studies with several GPCRs have suggested that beta-arrestins play an important role as signal transducers by scaffolding signaling molecules such as ERK1/2. This function of the beta-arrestins was not discovered until several years after their role in desensitization and endocytosis had been reported. In this study, we investigated the role of the beta-arrestins in the activation of ERK1/2 and receptor endocytosis. We took advantage of previously described mutants of FPR that have defects in G(i) coupling or beta-arrestin recruitment. The results obtained with the mutant FPRs, as well as experiments using an inhibitor of G(i) and cells overexpressing beta-arrestin2, showed that activation of ERK1/2 takes place through G(i) and is not affected by beta-arrestins. However, overexpression of beta-arrestin2 does enhance FPR sequestration from the cell surface, suggesting a role in desensitization, as shown for many other GPCRs. Experiments with CHO C5aR cells showed similar sensitivity to the G(i) inhibitor as CHO FPR cells, suggesting that the predominant activation of ERK1/2 through G protein may be a common characteristic among chemoattractant receptors.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Formyl Peptide/metabolism , Animals , Arrestins/metabolism , CHO Cells , Cricetinae , Cricetulus , Endocytosis/drug effects , Enzyme Activation/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , Humans , Ligands , Mice , Mutant Proteins/metabolism , NIH 3T3 Cells , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Receptor, Anaphylatoxin C5a/metabolism , Time Factors , Transfection , beta-Arrestins
16.
J Immunol ; 179(4): 2520-31, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17675514

ABSTRACT

The N-formyl peptide receptor (FPR), a G protein-coupled receptor that binds proinflammatory chemoattractant peptides, serves as a model receptor for leukocyte chemotaxis. Recombinant histidine-tagged FPR (rHis-FPR) was purified in lysophosphatidyl glycerol (LPG) by Ni(2+)-NTA agarose chromatography to >95% purity with high yield. MALDI-TOF mass analysis (>36% sequence coverage) and immunoblotting confirmed the identity as FPR. The rHis-FPR served as an immunogen for the production of 2 mAbs, NFPR1 and NFPR2, that epitope map to the FPR C-terminal tail sequences, 305-GQDFRERLI-313 and 337-NSTLPSAEVE-346, respectively. Both mAbs specifically immunoblotted rHis-FPR and recombinant FPR (rFPR) expressed in Chinese hamster ovary cells. NFPR1 also recognized recombinant FPRL1, specifically expressed in mouse L fibroblasts. In human neutrophil membranes, both Abs labeled a 45-75 kDa species (peak M(r) approximately 60 kDa) localized primarily in the plasma membrane with a minor component in the lactoferrin-enriched intracellular fractions, consistent with FPR size and localization. NFPR1 also recognized a band of M(r) approximately 40 kDa localized, in equal proportions to the plasma membrane and lactoferrin-enriched fractions, consistent with FPRL1 size and localization. Only NFPR2 was capable of immunoprecipitation of rFPR in detergent extracts. The recognition of rFPR by NFPR2 is lost after exposure of cellular rFPR to f-Met-Leu-Phe (fMLF) and regained after alkaline phosphatase treatment of rFPR-bearing membranes. In neutrophils, NFPR2 immunofluorescence was lost upon fMLF stimulation. Immunoblotting approximately 60 kDa species, after phosphatase treatment of fMLF-stimulated neutrophil membranes, was also enhanced. We conclude that the region 337-346 of FPR becomes phosphorylated after fMLF activation of rFPR-expressing Chinese hamster ovary cells and neutrophils.


Subject(s)
Antibodies, Monoclonal/chemistry , Epitopes/chemistry , Neutrophils/chemistry , Protein Processing, Post-Translational , Receptors, Formyl Peptide/chemistry , Animals , Antibodies, Monoclonal/immunology , CHO Cells , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/immunology , Cell Membrane/metabolism , Chemotaxis/drug effects , Chemotaxis/genetics , Chemotaxis/immunology , Chromatography, Affinity , Cricetinae , Cricetulus , Epitope Mapping , Epitopes/genetics , Epitopes/immunology , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression , Humans , Lactoferrin/chemistry , Lactoferrin/genetics , Lactoferrin/immunology , Lactoferrin/metabolism , Lysophospholipids/chemistry , Mice , Models, Immunological , N-Formylmethionine Leucyl-Phenylalanine/analogs & derivatives , N-Formylmethionine Leucyl-Phenylalanine/chemistry , N-Formylmethionine Leucyl-Phenylalanine/immunology , N-Formylmethionine Leucyl-Phenylalanine/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/immunology , Neutrophils/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/immunology , Protein Structure, Tertiary/genetics , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/isolation & purification , Receptors, Formyl Peptide/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spodoptera
17.
Cell Signal ; 17(10): 1300-11, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16038804

ABSTRACT

G protein-coupled receptors (GPCRs) transmit diverse cellular signals in response to a large number of stimuli such as chemoattractants, lipids, neurotransmitters, odorants and light. The classical signaling pathway is through heterotrimeric G proteins, but GPCRs can also transmit signals through mechanisms that are not dependent on G proteins. In mammalian cells, the key component for this type of signaling is the family of scaffolding molecules called beta-arrestins. They can function as scaffolds for activation of mitogen-activated protein kinases, including extracellular signal-regulated kinases 1 and 2 (ERK1/2). In this study we examined the role of G protein and beta-arrestin in formyl peptide receptor (FPR)-mediated activation of chemotaxis, receptor endocytosis and ERK1/2 activation using wild type and mutant receptors. Our findings suggest that, unlike certain other GPCRs that can activate ERK1/2 without the involvement of G protein, FPR requires signaling through a G protein-mediated pathway. Previous observations have shown that ERK1/2, activated through G protein, translocates to the nucleus where it stimulates transcription factors. In contrast, the scaffolding protein beta-arrestin retains the activated ERK1/2 in the cytoplasm to allow phosphorylation of cytoplasmic targets. Our experimental data show that both wild-type FPR and a mutant FPR, defective in beta-arrestin binding, induce nuclear translocation of activated ERK1/2 with similar ligand concentration dependence as seen for activation of cytosolic ERK1/2. We propose that FPR-mediated activation of ERK1/2 takes place primarily through G protein and is physiologically important to ensure transcriptional activation of myeloid immunomodulators, such as cytokines.


Subject(s)
Arrestins/metabolism , GTP-Binding Proteins/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Formyl Peptide/physiology , Animals , CHO Cells , Calcium/metabolism , Calcium Signaling/physiology , Cell Membrane/metabolism , Cell Nucleus/metabolism , Chemotaxis/drug effects , Chemotaxis/genetics , Chemotaxis/physiology , Cricetinae , Cricetulus , Cytosol/metabolism , Endocytosis , Enzyme Activation , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Mutation/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Receptors, Formyl Peptide/genetics , Signal Transduction/physiology , Transfection , beta-Arrestins
18.
Traffic ; 6(2): 100-15, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15634211

ABSTRACT

Two chemoattractant receptors, C5aR (the complement fragment C5a receptor) and FPR (the N-formyl peptide receptor), are involved in neutrophil activation at sites of inflammation. In this study, we found major differences in the intracellular trafficking of the receptors in transfected Chinese hamster ovary (CHO) cells. Western blot analysis showed that FPR was stable during a 3 h stimulation with ligand, but C5aR was reduced in quantity by 50%. Not all C5aR was targeted directly for degradation however; a small, but visible fraction of the receptor became re-phosphorylated upon subsequent addition of ligand, suggesting that some of the receptor had cycled to the cell surface. Light membrane fractions isolated from activated cells showed C5aR distribution at the bottom of a glycerol gradient, colocalizing with the main distribution of the late endosomal/lysosomal marker LAMP2, whereas FPR was found at the bottom of the gradient as well as in the middle of the gradient, where it cofractionated with the early/sorting endosomal marker Rab5. Using fluorescence microscopy, we observed ligand-dependent redistribution of C5aR-EGFP from the plasma membrane to LAMP2-positive compartments, whereas FPR-EGFP showed significant colocalization with the early/sorting endosomes. Analysis of endogenous C5aR and FPR in neutrophils revealed a pattern similar to the CHO transfectants: C5aR underwent degradation after prolonged ligand stimulation, while FPR did not. Finally, we confirmed the down-regulation of C5aR in a functional assay by showing reduced chemotaxis toward C5a in both CHO transfectants and neutrophils after preincubation with C5a. A similar decrease in FPR-mediated chemotaxis was not observed.


Subject(s)
Endocytosis , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Formyl Peptide/metabolism , Animals , Blotting, Western , CHO Cells , Chemotaxis/drug effects , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Down-Regulation , Fluorescent Antibody Technique, Indirect , Green Fluorescent Proteins/metabolism , Ligands , Microscopy, Confocal , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/physiology
19.
J Immunol ; 171(6): 3187-93, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12960347

ABSTRACT

A large number of G protein-coupled receptors have been shown to form homodimers based on a number of different techniques such as receptor coimmunoprecipitation, cross-linking, and fluorescence resonance energy transfer. In addition, functional assays of cells coexpressing a mutant receptor with a wild-type receptor have shown receptor phenotypes that can best be explained through dimerization. We asked whether the human neutrophil N-formyl peptide receptor (FPR) forms dimers in Chinese hamster ovary cells by coexpressing wild-type FPR with one of two mutants: D71A, which is uncoupled from G protein, and N297A, which has a defect in receptor phosphorylation and endocytosis. Experiments measuring chemotaxis, ligand-induced release of intracellular calcium, and p42/44 mitogen-activated protein kinase activation did not show an inhibitory effect of the coexpressed FPR D71A mutant. Coexpressed wild-type receptor was efficiently internalized, but failed to correct the endocytosis defects of the D71A and the N297A mutants. To explore the possibility that the mutations themselves prevented dimerization, we examined the coimmunoprecipitation of differentially epitope-tagged FPR. Immunoprecipitation of hemagglutinin-tagged FPR failed to coimmunoprecipitate coexpressed c-myc-tagged FPR and vice versa. Together, these data suggest that, unlike many other G protein-coupled receptors, FPR does not form homodimers.


Subject(s)
Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/physiology , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Asparagine/genetics , Aspartic Acid/genetics , CHO Cells , Calcium/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Chemotaxis, Leukocyte/genetics , Cricetinae , Dimerization , Endocytosis/genetics , Enzyme Activation/genetics , Epitopes/metabolism , Humans , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Oligopeptides , Peptides/metabolism , Precipitin Tests , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/genetics , Transfection
20.
J Periodontol ; 74(4): 475-84, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12747452

ABSTRACT

BACKGROUND: Localized aggressive periodontitis (LAgP) is a disease characterized by rapid loss of alveolar bone in teeth of otherwise healthy patients. Neutrophils from LAgP patients have been shown to exhibit diminished chemotaxis and low levels of formyl peptide receptor (FPR) surface expression. A recent study has associated LAgP with 2 polymorphisms in the FPR: 110Phe-->Ser and 126Cys-->Trp. METHODS: We transfected Chinese hamster ovary cells with wtFPR, FPR-110Phe-->Ser, FPR-126Cys-->Trp, or FPR-110Phe-->Ala and determined their surface expression of FPR, their ligand binding affinity, their G-protein coupling, and their chemotaxis toward N-formyl-methionyl-leucyl-phenylalanine (FMLP). RESULTS: FPR-110Phe-->Ser mutants failed to show any significant surface expression or chemotaxis toward FMLP. FPR-126Cys-->Trp mutants exhibited slightly lower than normal binding affinity, markedly lower G-protein coupling response, and markedly lower chemotaxis toward FMLP than that observed with wtFPR. We also analyzed another FPR-Phe110 mutant, FPR-110Phe-->Ala, to ascertain what the effect of mutating this residue might be in a mutant that could be expressed on the cell surface. The FPR-110Phe-->Ala mutant demonstrated markedly lower surface expression, normal ligand binding affinity, markedly lower G-protein coupling, and markedly lower chemotaxis toward FMLP. CONCLUSIONS: Our data substantiate the hypothesis that the chemotactic defects observed in LAgP patients are due at least in part to molecular alterations in the FPR. The FPR-110Phe-->Ser polymorphism appears to be more defective than the FPR-126Cys-->Trp polymorphism, indicating that patients with the former polymorphism might be expected to exhibit a more severe form of aggressive periodontitis.


Subject(s)
Aggressive Periodontitis/genetics , Chemotaxis/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Aggressive Periodontitis/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Cysteine/genetics , Models, Chemical , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenylalanine/genetics , Polymorphism, Genetic , Protein Binding , Protein Structure, Secondary , Receptors, Formyl Peptide , Receptors, Immunologic/chemistry , Receptors, Peptide/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...