Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Virol ; 92(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-29997213

ABSTRACT

Like many other large double-stranded DNA (dsDNA) viruses, herpesviruses are known to capture host genes to evade host defenses. Little is known about the detailed natural history of such genes, nor do we fully understand their evolutionary dynamics. A major obstacle is that they are often highly divergent, maintaining very low sequence similarity to host homologs. Here we use the herpesvirus genus Rhadinovirus as a model system to develop an analytical approach that combines complementary evolutionary and bioinformatic techniques, offering results that are both detailed and robust for a range of genes. Using a systematic phylogenetic strategy, we identify the original host lineage of viral genes with high confidence. We show that although host immunomodulatory genes evolve rapidly compared to other host genes, they undergo a clear increase in purifying selection once captured by a virus. To characterize this shift in detail, we developed a novel technique to identify changes in selection pressure that can be attributable to particular domains. These findings will inform us on how viruses develop strategies to evade the immune system, and our synthesis of techniques can be reapplied to other viruses or biological systems with similar analytical challenges.IMPORTANCE Viruses and hosts have been shown to capture genes from one another as part of the evolutionary arms race. Such genes offer a natural experiment on the effects of evolutionary pressure, since the same gene exists in vastly different selective environments. However, sequences of viral homologs often bear little similarity to the original sequence, complicating the reconstruction of their shared evolutionary history with host counterparts. In this study, we use a genus of herpesviruses as a model system to comprehensively investigate the evolution of host-derived viral genes, using a synthesis of genomics, phylogenetics, selection analysis, and nucleotide and amino acid modeling.


Subject(s)
Genes, Viral/immunology , Histocompatibility Antigens Class I/genetics , Host-Pathogen Interactions , Rhadinovirus/genetics , Selection, Genetic , Viral Proteins/genetics , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/immunology , Atelinae/virology , Biological Evolution , CD59 Antigens/chemistry , CD59 Antigens/genetics , CD59 Antigens/immunology , Callithrix/virology , Chemokine CCL3/chemistry , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Computational Biology , Gene Expression Regulation , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Interleukin-17/chemistry , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Models, Molecular , Phylogeny , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Rats , Rhadinovirus/chemistry , Rhadinovirus/immunology , Saimiri/virology , Viral Proteins/chemistry , Viral Proteins/immunology
2.
Proc Natl Acad Sci U S A ; 113(18): 5000-5, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27091995

ABSTRACT

CC chemokine ligand 5 (CCL5) and CCL3 are critical for immune surveillance and inflammation. Consequently, they are linked to the pathogenesis of many inflammatory conditions and are therapeutic targets. Oligomerization and glycosaminoglycan (GAG) binding of CCL5 and CCL3 are vital for the functions of these chemokines. Our structural and biophysical analyses of human CCL5 reveal that CCL5 oligomerization is a polymerization process in which CCL5 forms rod-shaped, double-helical oligomers. This CCL5 structure explains mutational data and offers a unified mechanism for CCL3, CCL4, and CCL5 assembly into high-molecular-weight, polydisperse oligomers. A conserved, positively charged BBXB motif is key for the binding of CC chemokines to GAG. However, this motif is partially buried when CCL3, CCL4, and CCL5 are oligomerized; thus, the mechanism by which GAG binds these chemokine oligomers has been elusive. Our structures of GAG-bound CCL5 and CCL3 oligomers reveal that these chemokine oligomers have distinct GAG-binding mechanisms. The CCL5 oligomer uses another positively charged and fully exposed motif, KKWVR, in GAG binding. However, residues from two partially buried BBXB motifs along with other residues combine to form a GAG-binding groove in the CCL3 oligomer. The N termini of CC chemokines are shown to be involved in receptor binding and oligomerization. We also report an alternative CCL3 oligomer structure that reveals how conformational changes in CCL3 N termini profoundly alter its surface properties and dimer-dimer interactions to affect GAG binding and oligomerization. Such complexity in oligomerization and GAG binding enables intricate, physiologically relevant regulation of CC chemokine functions.


Subject(s)
Chemokine CCL3/chemistry , Chemokine CCL3/ultrastructure , Chemokine CCL5/chemistry , Chemokine CCL5/ultrastructure , Glycosaminoglycans/chemistry , Binding Sites , Dimerization , Humans , Protein Binding , Protein Conformation , Structure-Activity Relationship
3.
Stem Cells Dev ; 25(8): 575-85, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26943017

ABSTRACT

Dipeptidyl peptidase 4 (DPP4)/CD26 truncates certain proteins, and this posttranslational modification can influence their activity. Truncated (T) colony-stimulating factors (CSFs) are decreased in potency for stimulating proliferation of hematopoietic progenitor cells (HPCs). T-CXCL12, a modified chemokine, is inactive as an HPC chemotactic, survival, and enhancing factor for replating or ex-vivo expansion of HPCs. Moreover, T-CSFs and T-CXCL12 specifically downmodulates the positively acting effects of their own full-length molecule. Other chemokines have DPP4 truncation sites. In the present study, we evaluated effects of DPP4 inhibition (by Diprotin A) or gene deletion of HPC on chemokine inhibition of multicytokine-stimulated HPC, and on chemokine-enhancing effects on single CSF-stimulated HPC proliferation, as well as effects of DPP4 treatment of a number of chemokines. Myelosuppressive effects of chemokines with, but not without, a DPP4 truncation site were greatly enhanced in inhibitory potency by pretreating target bone marrow (BM) cells with Diprotin A, or by assaying their activity on dpp4/cd26(-/-) BM cells. DPP4 treatment of myelosuppressive chemokines containing a DPP4 truncation site produced a nonmyelosuppressive molecule, but one which had the capacity to block suppression by that unmodified chemokine both in vitro and in vivo. Additionally, DPP4 treatment ablated the single cytokine-stimulated HPC-enhancing activity of CCL3/MIP-1α and CCL4/MIP-1ß, and blocked the enhancing activity of each unmodified molecule, in vitro and in vivo. These results highlight the functional posttranslational modulating effects of DPP4 on chemokine activities, and information offering additional biological insight into chemokine regulation of hematopoiesis.


Subject(s)
Chemokine CCL3/physiology , Chemokine CCL4/physiology , Dipeptidyl Peptidase 4/physiology , Animals , Cell Proliferation , Chemokine CCL3/chemistry , Chemokine CCL4/chemistry , Dipeptidyl Peptidase 4/chemistry , Female , Hematopoiesis , Hematopoietic Stem Cells/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational , Proteolysis
4.
Structure ; 24(1): 57-69, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26671708

ABSTRACT

A wide variety of pathogens targets chemokine signaling networks in order to disrupt host immune surveillance and defense. Here, we report a structural and mutational analysis of rodent herpesvirus Peru encoded R17, a potent chemokine inhibitor that sequesters CC and C chemokines with high affinity. R17 consists of a pair of ß-sandwich domains linked together by a bridging sheet, which form an acidic binding cleft for the chemokine CCL3 on the opposite face of a basic surface cluster that binds glycosaminoglycans. R17 promiscuously engages chemokines primarily through the same N-loop determinants used for host receptor recognition while residues located in the chemokine 40s loop drive kinetically stable complex formation. The core fold adopted by R17 is unexpectedly similar to that of the M3 chemokine decoy receptor encoded by MHV-68, although, strikingly, neither the location of ligand engagement nor the stoichiometry of binding is conserved, suggesting that their functions evolved independently.


Subject(s)
Chemokine CCL3/chemistry , Evolution, Molecular , Viral Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Conserved Sequence , Herpesviridae/chemistry , Mice , Molecular Sequence Data , Protein Binding , Viral Proteins/genetics , Viral Proteins/metabolism
5.
J Biomol NMR ; 63(4): 333-340, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26472202

ABSTRACT

C-C chemokine receptor 1 (CCR1) and CCR5 are involved in various inflammation and immune responses, and regulate the progression of the autoimmune diseases differently. However, the number of residues identified at the binding interface was not sufficient to clarify the differences in the CCR1- and CCR5-binding modes to MIP-1α, because the NMR measurement time for CCR1 and CCR5 samples was limited to 24 h, due to their low stability. Here we applied a recently developed NMR spectra reconstruction method, Conservation of experimental data in ANAlysis of FOuRier, to the amide-directed transferred cross-saturation experiments of chemokine receptors, CCR1 and CCR5, embedded in lipid bilayers of the reconstituted high density lipoprotein, and MIP-1α. Our experiments revealed that the residues on the N-loop and ß-sheets of MIP-1α are close to both CCR1 and CCR5, and those in the C-terminal helix region are close to CCR5. These results suggest that the genetic influence of the single nucleotide polymorphisms of MIP-1α that accompany substitution of residues in the C-terminal helix region, E57 and V63, would provide clues toward elucidating how the CCR5-MIP-1α interaction affects the progress of autoimmune diseases.


Subject(s)
Chemokine CCL3/chemistry , Receptors, CCR1/chemistry , Receptors, CCR5/chemistry , Animals , Binding Sites , Cell Line , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Spodoptera
6.
J Mol Biol ; 427(6 Pt B): 1345-1358, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25636406

ABSTRACT

CC chemokine ligands (CCLs) are 8- to 14-kDa signaling proteins involved in diverse immune functions. While CCLs share similar tertiary structures, oligomerization produces highly diverse quaternary structures that protect chemokines from proteolytic degradation and modulate their functions. CCL18 is closely related to CCL3 and CCL4 with respect to both protein sequence and genomic location, yet CCL18 has distinct biochemical and biophysical properties. Here, we report a crystal structure of human CCL18 and its oligomerization states in solution based on crystallographic and small-angle X-ray scattering analyses. Our data show that CCL18 adopts an α-helical conformation at its N-terminus that weakens its dimerization, explaining CCL18's preference for the monomeric state. Multiple contacts between monomers allow CCL18 to reversibly form a unique open-ended oligomer different from those of CCL3, CCL4, and CCL5. Furthermore, these differences hinge on proline 8, which is conserved in CCL3 and CCL4 but is replaced by lysine in human CCL18. Our structural analyses suggest that a mutation of proline 8 to alanine stabilizes a type 1 ß-turn at the N-terminus of CCL4 to prevent dimerization but prevents dimers from making key contacts with each other in CCL3. Thus, the P8A mutation induces depolymerization of CCL3 and CCL4 by distinct mechanisms. Finally, we used structural, biochemical, and functional analyses to unravel why insulin-degrading enzyme degrades CCL3 and CCL4 but not CCL18. Our results elucidate the molecular basis for the oligomerization of three closely related CC chemokines and suggest how oligomerization shapes CCL chemokine function.


Subject(s)
Chemokine CCL3/chemistry , Chemokine CCL4/chemistry , Chemokines, CC/metabolism , Insulysin/metabolism , Amino Acid Sequence , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Chemokine CCL4/genetics , Chemokine CCL4/metabolism , Chemokines, CC/chemistry , Chemokines, CC/genetics , Chemotaxis , Crystallography, X-Ray , Humans , Insulysin/chemistry , Insulysin/genetics , Molecular Sequence Data , Mutation/genetics , Protein Structure, Quaternary , Scattering, Small Angle , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
J Biol Chem ; 289(46): 31846-31855, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25266725

ABSTRACT

To elucidate the ligand-binding surface of the CC chemokine-binding proteins Evasin-1 and Evasin-4, produced by the tick Rhipicephalus sanguineus, we sought to identify the key determinants responsible for their different chemokine selectivities by expressing Evasin mutants using phage display. We first designed alanine mutants based on the Evasin-1·CCL3 complex structure and an in silico model of Evasin-4 bound to CCL3. The mutants were displayed on M13 phage particles, and binding to chemokine was assessed by ELISA. Selected variants were then produced as purified proteins and characterized by surface plasmon resonance analysis and inhibition of chemotaxis. The method was validated by confirming the importance of Phe-14 and Trp-89 to the inhibitory properties of Evasin-1 and led to the identification of a third crucial residue, Asn-88. Two amino acids, Glu-16 and Tyr-19, were identified as key residues for binding and inhibition of Evasin-4. In a parallel approach, we identified one clone (Y28Q/N60D) that showed a clear reduction in binding to CCL3, CCL5, and CCL8. It therefore appears that Evasin-1 and -4 use different pharmacophores to bind CC chemokines, with the principal binding occurring through the C terminus of Evasin-1, but through the N-terminal region of Evasin-4. However, both proteins appear to target chemokine N termini, presumably because these domains are key to receptor signaling. The results also suggest that phage display may offer a useful approach for rapid investigation of the pharmacophores of small inhibitory binding proteins.


Subject(s)
Chemokines, CC/chemistry , Receptors, Chemokine/chemistry , Alanine/chemistry , Amino Acid Sequence , Animals , Cell Movement , Chemokine CCL3/chemistry , Chemokine CCL5/chemistry , Chemokine CCL5/genetics , Chemokine CCL8/chemistry , Chemotaxis , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Glycosylation , HEK293 Cells , Humans , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Library , Protein Binding , Protein Structure, Tertiary , Rhipicephalus sanguineus , Sequence Homology, Amino Acid , Surface Plasmon Resonance
8.
Cytokine ; 55(2): 168-73, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21632261

ABSTRACT

Chemokines and their receptors control cell migration associated with routine immune surveillance, inflammation and development. They are also implicated in a large number of inflammatory diseases, cancer and HIV. Here we describe a rapid and efficient way to express and purify milligram quantities of multiple chemokine ligands (CCL7/MCP-3, CCL14/HCC-1, CCL3/MIP-1α and CXCL8/IL-8) containing C-terminal modifications to enable coupling to fluorescent dyes or small molecules such as biotin, in vitro. These labeled chemokines display wild-type behavior in both receptor binding and calcium mobilization assays. The ability to rapidly and inexpensively produce labeled chemokines opens the way for their use in many applications, including non-traditional chemokine-receptor interaction studies, both on intact cells and with purified receptor reconstituted in artificial membranes in vitro. Furthermore, the ability to immobilize chemokines to obtain ligand affinity columns aids in efforts to purify chemokine receptors for structural and biophysical studies, by facilitating the separation of functional proteins from their non-functional counterparts.


Subject(s)
Chemokines/chemistry , Chemokines/isolation & purification , Chromatography, Affinity/methods , Biotin/chemistry , Biotin/metabolism , Chemokine CCL3/chemistry , Chemokine CCL3/genetics , Chemokine CCL3/isolation & purification , Chemokine CCL7/chemistry , Chemokine CCL7/genetics , Chemokine CCL7/isolation & purification , Chemokines/genetics , Chemokines, CC/chemistry , Chemokines, CC/genetics , Chemokines, CC/isolation & purification , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Interleukin-8/chemistry , Interleukin-8/genetics , Interleukin-8/isolation & purification , Ligands , Radioligand Assay , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
EMBO J ; 29(23): 3952-66, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20959807

ABSTRACT

Macrophage inflammatory protein-1 (MIP-1), MIP-1α (CCL3) and MIP-1ß (CCL4) are chemokines crucial for immune responses towards infection and inflammation. Both MIP-1α and MIP-1ß form high-molecular-weight aggregates. Our crystal structures reveal that MIP-1 aggregation is a polymerization process and human MIP-1α and MIP-1ß 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.


Subject(s)
Chemokine CCL3/chemistry , Chemokine CCL3/metabolism , Chemokine CCL4/chemistry , Chemokine CCL4/metabolism , Insulysin/metabolism , 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
10.
J Am Chem Soc ; 132(19): 6768-77, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20423099

ABSTRACT

CC-chemokine receptor 5 (CCR5) belongs to the G protein-coupled receptor (GPCR) family and plays important roles in the inflammatory response. In addition, its ligands inhibit the HIV infection. Structural analyses of CCR5 have been hampered by its instability in the detergent-solubilized form. Here, CCR5 was reconstituted into high density lipoprotein (rHDL), which enabled CCR5 to maintain its functions for >24 h and to be suitable for structural analyses. By applying the methyl-directed transferred cross-saturation (TCS) method to the complex between rHDL-reconstituted CCR5 and its ligand MIP-1alpha, we demonstrated that valine 59 and valine 63 of MIP-1alpha are in close proximity to CCR5 in the complex. Furthermore, these results suggest that the protective influence on HIV-1 infection of a SNP of MIP-1alpha is due to its change of affinity for CCR5. This method will be useful for investigating the various and complex signaling mediated by GPCR, and will also provide structural information about the interactions of other GPCRs with lipids, ligands, G-proteins, and effector molecules.


Subject(s)
Lipid Bilayers/metabolism , Nuclear Magnetic Resonance, Biomolecular , Receptors, CCR5/chemistry , Receptors, CCR5/metabolism , Amino Acid Sequence , Animals , Cell Line , Chemokine CCL3/chemistry , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Genetic Variation , Humans , Ligands , Lipid Bilayers/chemistry , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Maltose/analogs & derivatives , Maltose/chemistry , Micelles , Models, Molecular , Protein Binding , Protein Conformation , Protein Stability , Solutions
11.
PLoS One ; 4(12): e8514, 2009 Dec 30.
Article in English | MEDLINE | ID: mdl-20041127

ABSTRACT

BACKGROUND: Chemokines are a subset of cytokines responsible for controlling the cellular migration of inflammatory cells through interaction with seven transmembrane G protein-coupled receptors. The blocking of a chemokine-receptor interaction results in a reduced inflammatory response, and represents a possible anti-inflammatory strategy, a strategy that is already employed by some virus and parasites. Anti-chemokine activity has been described in the extracts of tick salivary glands, and we have recently described the cloning and characterization of such chemokine binding proteins from the salivary glands, which we have named Evasins. METHODOLOGY/PRINCIPAL FINDINGS: We have solved the structure of Evasin-1, a very small and highly selective chemokine-binding protein, by x-ray crystallography and report that the structure is novel, with no obvious similarity to the previously described structures of viral chemokine binding proteins. Moreover it does not possess a known fold. We have also solved the structure of the complex of Evasin-1 and its high affinity ligand, CCL3. The complex is a 1:1 heterodimer in which the N-terminal region of CCL3 forms numerous contacts with Evasin-1, including prominent pi-pi interactions between residues Trp89 and Phe14 of the binding protein and Phe29 and Phe13 of the chemokine. CONCLUSIONS/SIGNIFICANCE: However, these interactions do not appear to be crucial for the selectivity of the binding protein, since these residues are found in CCL5, which is not a ligand for Evasin-1. The selectivity of the interaction would appear to lie in the N-terminal residues of the chemokine, which form the "address" whereas the hydrophobic interactions in the rest of the complex would serve primarily to stabilize the complex. A thorough understanding of the binding mode of this small protein, and its other family members, could be very informative in the design of potent neutralizing molecules of pro-inflammatory mediators of the immune system, such as chemokines.


Subject(s)
Chemokine CCL3/chemistry , Chemokine CCL3/metabolism , Receptors, Chemokine/chemistry , Receptors, Chemokine/metabolism , Rhipicephalus/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , Glycosylation , Humans , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Static Electricity , Structure-Activity Relationship
12.
Protein Sci ; 18(5): 1115-20, 2009 May.
Article in English | MEDLINE | ID: mdl-19388076

ABSTRACT

Protein aggregation is an essential molecular event in a wide variety of biological situations, and is a causal factor in several degenerative diseases. The aggregation of proteins also frequently hampers structural biological analyses, such as solution NMR studies. Therefore, precise detection and characterization of protein aggregation are of crucial importance for various research fields. In this study, we demonstrate that fluorescence correlation spectroscopy (FCS) using a single-molecule fluorescence detection system enables the detection of otherwise invisible aggregation of proteins at higher protein concentrations, which are suitable for structural biological experiments, and consumes relatively small amounts of protein over a short measurement time. Furthermore, utilizing FCS, we established a method for high-throughput screening of protein aggregation and optimal solution conditions for structural biological experiments.


Subject(s)
Proteins , Proteomics/methods , Spectrometry, Fluorescence/methods , Chemokine CCL3/chemistry , Chemokine CCL3/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proteins/chemistry , Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 105(39): 14826-31, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18815363

ABSTRACT

G protein-coupled receptors (GPCRs) have a key role in many biological processes and are important drug targets for many human diseases. Therefore, understanding the molecular interactions between GPCRs and their ligands would improve drug design. Here, we describe an approach that allows the rapid identification of functional agonists expressed in bacteria. Transgenic Caenorhabditis elegans expressing the human chemokine receptor 5 (CCR5) in nociceptive neurons show avoidance behavior on encounter with the ligand MIP-1alpha and avoid feeding on Escherichia coli expressing MIP-1alpha compared with control bacteria. This system allows a simple activity screen, based on the distribution of transgenic worms in a binary food-choice assay, without a requirement for protein purification or tagging. By using this approach, a library of 68 MIP-1alpha variants was screened, and 13 critical agonist residues involved in CCR5 activation were identified, four of which (T8, A9, N22, and A25) have not been described previously, to our knowledge. Identified residues were subsequently validated in receptor binding assays and by calcium flux assays in mammalian cells. This approach serves not only for structure/function studies as demonstrated, but may be used to facilitate the discovery of agonists within bacterial libraries.


Subject(s)
CCR5 Receptor Antagonists , Caenorhabditis elegans/physiology , Chemokine CCL3/biosynthesis , Escherichia coli/metabolism , Feeding Behavior , Receptors, G-Protein-Coupled/agonists , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Chemokine CCL3/chemistry , Chemokine CCL3/genetics , Escherichia coli/genetics , Escherichia coli/isolation & purification , Gene Library , Humans , Ligands , Neurons/metabolism , Protein Binding , Protein Conformation , Receptors, CCR5/genetics , Receptors, G-Protein-Coupled/genetics , Transgenes
14.
J Biol Chem ; 283(34): 23121-8, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18559339

ABSTRACT

7 transmembrane-spanning (7TM) chemokine receptors having multiple endogenous ligands offer special opportunities to understand the molecular basis for allosteric mechanisms. Thus, CC-chemokine receptor 1 (CCR1) binds CC-chemokine 3 and 5 (CCL3 and CCL5) with K(d) values of 7.3 and 0.16 nm, respectively, as determined in homologous competition binding assays. However, CCL5 appears to have a >10,000-fold lower affinity in competition against (125)I-CCL3. Mutational mapping revealed that CCL3 and CCL5 both are strongly affected by systematic truncations of the N-terminal extension, whereas only CCL5 and not CCL3 activation is affected by substitutions in the main ligand binding pocket including the conserved GluVII:06 anchor point. A series of metal ion chelator complexes were found to act as full agonists on CCR1 and to be critically affected by the same substitutions in the main ligand binding pocket as CCL5 but not by mutations in the extracellular domain. In agreement with the overlapping binding sites, the small non-peptide agonists displaced radiolabeled CCL5 with high affinity. Interestingly, the same compounds acted as allosteric enhancers of the binding of CCL3, with which they did not overlap in binding site, leading to an increased B(max) and affinity of this chemokine mainly due to an increased association rate. It is concluded that a small molecule agonist through binding deep in the main ligand binding pocket can act as an allosteric enhancer for one endogenous chemokine and at the same time as a competitive blocker of the binding of another endogenous chemokine.


Subject(s)
Gene Expression Regulation , Receptors, CCR1/physiology , Allosteric Site , Amino Acid Sequence , Animals , COS Cells , Chemokine CCL3/chemistry , Chemokine CCL5/chemistry , Chlorocebus aethiops , Humans , Kinetics , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , Receptors, CCR1/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...