Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Cell ; 187(17): 4751-4769.e25, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39089252

ABSTRACT

The Duffy antigen receptor is a seven-transmembrane (7TM) protein expressed primarily at the surface of red blood cells and displays strikingly promiscuous binding to multiple inflammatory and homeostatic chemokines. It serves as the basis of the Duffy blood group system in humans and also acts as the primary attachment site for malarial parasite Plasmodium vivax and pore-forming toxins secreted by Staphylococcus aureus. Here, we comprehensively profile transducer coupling of this receptor, discover potential non-canonical signaling pathways, and determine the cryoelectron microscopy (cryo-EM) structure in complex with the chemokine CCL7. The structure reveals a distinct binding mode of chemokines, as reflected by relatively superficial binding and a partially formed orthosteric binding pocket. We also observe a dramatic shortening of TM5 and 6 on the intracellular side, which precludes the formation of the docking site for canonical signal transducers, thereby providing a possible explanation for the distinct pharmacological and functional phenotype of this receptor.


Subject(s)
Cryoelectron Microscopy , Duffy Blood-Group System , Receptors, Cell Surface , Humans , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/chemistry , Duffy Blood-Group System/metabolism , Duffy Blood-Group System/chemistry , Signal Transduction , Binding Sites , Chemokines/metabolism , Chemokines/chemistry , Protein Binding
2.
ACS Appl Mater Interfaces ; 16(29): 37390-37400, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39007843

ABSTRACT

Staphylococcus aureus (S. aureus) is an opportunistic infectious pathogen, which causes a high mortality rate during bloodstream infections. The early detection of virulent strains in patients' blood samples is of medical interest for rapid diagnosis. The main virulent factors identified in patient isolates include leukocidins that bind to specific membrane receptors and lyse immune cells and erythrocytes. Duffy antigen receptor for chemokines (DARC) on the surface of specific cells is a main target of leukocidins such as gamma-hemolysin AB (HlgAB) and leukocidin ED (LukED). Among them, HlgAB is a conserved and critical leukocidin that binds to DARC and forms pores on the cell membranes, leading to cell lysis. Current methods are based on ELISA or bacterial culture, which takes hours to days. For detecting HlgAB with faster response and higher sensitivity, we developed a biosensor that combines single-walled carbon nanotube field effect transistors (swCNT-FETs) with immobilized DARC receptors as biosensing elements. DARC was purified from a bacterial expression system and successfully reconstituted into nanodiscs that preserve binding capability for HlgAB. Dynamic light scattering (DLS) and scanning electron microscopy (SEM) showed an increase of the DARC-containing nanodisc size in the presence of HlgAB, indicating the formation of HlgAB prepore or pore complexes. We demonstrate that this sensor can specifically detect the leukocidins HlgA and HlgAB in a quantitative manner within the dynamic range of 1 fM to 100 pM with an LOD of 0.122 fM and an LOQ of 0.441 fM. The sensor was challenged with human serum spiked with HlgAB as simulated clinical samples. After dilution for decreasing nonspecific binding, it selectively detected the toxin with a similar detection range and apparent dissociation constant as in the buffer. This biosensor was demonstrated with remarkable sensitivity to detect HlgAB rapidly and has the potential as a tool for fundamental research and clinical applications, although this sensor cannot differentiate between HlgAB and LukED as both have the same receptor.


Subject(s)
Biosensing Techniques , Duffy Blood-Group System , Leukocidins , Staphylococcus aureus , Biosensing Techniques/methods , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/metabolism , Leukocidins/chemistry , Leukocidins/metabolism , Humans , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/chemistry , Nanotubes, Carbon/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism
3.
J Biol Chem ; 295(50): 17241-17250, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33051210

ABSTRACT

Leukocidin ED (LukED) is a pore-forming toxin produced by Staphylococcus aureus, which lyses host cells and promotes virulence of the bacteria. LukED enables S. aureus to acquire iron by lysing erythrocytes, which depends on targeting the host receptor Duffy antigen receptor for chemokines (DARC). The toxin also targets DARC on the endothelium, contributing to the lethality observed during bloodstream infection in mice. LukED is comprised of two monomers: LukE and LukD. LukE binds to DARC and facilitates hemolysis, but the closely related Panton-Valentine leukocidin S (LukS-PV) does not bind to DARC and is not hemolytic. The interaction of LukE with DARC and the role this plays in hemolysis are incompletely characterized. To determine the domain(s) of LukE that are critical for DARC binding, we studied the hemolytic function of LukE-LukS-PV chimeras, in which areas of sequence divergence (divergence regions, or DRs) were swapped between the toxins. We found that two regions of LukE's rim domain contribute to hemolysis, namely residues 57-75 (DR1) and residues 182-196 (DR4). Interestingly, LukE DR1 is sufficient to render LukS-PV capable of DARC binding and hemolysis. Further, LukE, by binding DARC through DR1, promotes the recruitment of LukD to erythrocytes, likely by facilitating LukED oligomer formation. Finally, we show that LukE targets murine Darc through DR1 in vivo to cause host lethality. These findings expand our biochemical understanding of the LukE-DARC interaction and the role that this toxin-receptor pair plays in S. aureus pathophysiology.


Subject(s)
Bacterial Proteins , Duffy Blood-Group System , Erythrocytes , Exotoxins , Hemolysin Proteins , Receptors, Cell Surface , Staphylococcus aureus , Animals , Humans , Mice , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/genetics , Duffy Blood-Group System/metabolism , Erythrocytes/chemistry , Erythrocytes/metabolism , Exotoxins/chemistry , Exotoxins/genetics , Exotoxins/metabolism , Protein Domains , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
4.
Blood ; 130(12): 1441-1444, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28754683

ABSTRACT

Plasmodium vivax is the most prevalent parasite species that causes malaria in humans and exclusively infects reticulocytes. Reticulocyte infection is facilitated by P vivax Duffy binding protein (DBP), which utilizes DARC (Duffy antigen receptor for chemokines) as an entry point. However, the selective tropism of P vivax for transferrin receptor (CD71)-positive reticulocytes remained unexplained, given the constitutive expression of DARC during reticulocyte maturation. CD71/RNA double staining of reticulocytes enriched from adult peripheral blood reveals 4 distinct reticulocyte populations: CD71high/RNAhigh (∼0.016%), CD71low/RNAhigh (∼0.059%), CD71neg/RNAhigh (∼0.37%), CD71neg/RNAlow (∼0.55%), and erythrocytes CD71neg/RNAneg (∼99%). We hypothesized that selective association of DBP with a small population of immature reticulocytes could explain the preference of P vivax for reticulocytes. Binding of specific monoclonal anti-DARC antibodies and recombinant DBP to CD71high/RNAhigh reticulocytes was significantly higher compared with other reticulocyte populations and erythrocytes. Interestingly, the total DARC protein throughout reticulocyte maturation was constant. The data suggest that selective exposure of the DBP binding site within DARC is key to the preferential binding of DBP to immature reticulocytes, which is the potential mechanism underlying the preferential infection of a reticulocyte subset by P vivax.


Subject(s)
Duffy Blood-Group System/chemistry , Duffy Blood-Group System/metabolism , Extracellular Space/chemistry , Plasmodium vivax/physiology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Reticulocytes/cytology , Reticulocytes/metabolism , Tropism/physiology , Antibody Specificity/immunology , Antigens, Protozoan/metabolism , Cell Differentiation , Erythrocytes/parasitology , Humans , Protein Domains , Protozoan Proteins/metabolism , Structure-Activity Relationship
5.
Postepy Hig Med Dosw (Online) ; 70: 143-61, 2016 Mar 04.
Article in Polish | MEDLINE | ID: mdl-26943312

ABSTRACT

Duffy (Fy) blood group antigens are located on seven-transmembrane glycoprotein expressed on erythrocytes and endothelial cells, which acts as atypical chemokine receptor (ACKR1) and malarial receptor. The biological role of the Duffy glycoprotein has not been explained yet. It is suggested that Duffy protein modulate the intensity of the inflammatory response. The Duffy blood group system consists of two major antigens, Fy(a) and Fy(b), encoded by two codominant alleles designated FY*A and FY*B which differ by a single nucleotide polymorphism (SNP) at position 125G>A of the FY gene that results in Gly42Asp amino acid change in the Fy(a) and Fy(b) antigens, respectively. The presence of antigen Fy(a) and/or Fy(b) on the erythrocytes determine three Duffy-positive phenotypes: Fy(a+b-), Fy(a-b+) and Fy(a+b+), identified in Caucasian population. The Duffy-negative phenotype Fy(a-b-), frequent in Africans, but very rare in Caucasians, is defined by the homozygous state of FY*B-33 alleles. The FY*B-33 allele is associated with a SNP -33T>C in the promoter region of the FY gene, which suppresses erythroid expression of this gene without affecting its expression in other tissues. The FY*X allele, found in Caucasians, is correlated with weak expression of Fy(b) antigen. Fy(x) antigen differs from the native Fy(b) by the Arg89Cys and Ala100Thr amino acid substitutions due to SNPs: 265C>T and 298G>A in FY*B allele. The frequency of the FY alleles shows marked geographic disparities, the FY*B-33 allele is predominant in Africans, the FY*B in Caucasians, while the FY*A allele is dominant in Asians and it is the most prevalent allele globally.


Subject(s)
Duffy Blood-Group System/chemistry , Duffy Blood-Group System/genetics , Alleles , Erythrocytes/metabolism , Homozygote , Humans , Phenotype , Polymorphism, Single Nucleotide
6.
PLoS One ; 9(8): e103262, 2014.
Article in English | MEDLINE | ID: mdl-25084090

ABSTRACT

The malaria parasite Plasmodium vivax is known to be majorly endemic to Asian and Latin American countries with no or very few reports of Africans infected with this parasite. Since the human Duffy antigens act as receptors for P. vivax to invade human RBCs and Africans are generally Duffy-negative, non-endemicity of P. vivax in Africa has been attributed to this fact. However, recent reports describing P. vivax infections in Duffy-negative Africans from West and Central parts of Africa have been surfaced including a recent report on P. vivax infection in native Cameroonians. In order to know if Cameroonians living in the southern regions are also susceptible to P. vivax infection, we collected finger-prick blood samples from 485 malarial symptomatic patients in five locations and followed PCR diagnostic assays with DNA sequencing of the 18S ribosomal RNA gene. Out of the 201 malaria positive cases detected, 193 were pure P. falciparum, six pure P. vivax and two mixed parasite infections (P. falciparum + P. vivax). The eight P. vivax infected samples (six single + two mixed) were further subjected to DNA sequencing of the P. vivax multidrug resistance 1 (pvmdr1) and the P.vivax circumsporozoite (pvcsp) genes. Alignment of the eight Cameroonian pvmdr1 sequences with the reference sequence showed high sequence similarities, reconfirming P. vivax infection in all the eight patients. DNA sequencing of the pvcsp gene indicated all the eight P. vivax to be of VK247 type. Interestingly, DNA sequencing of a part of the human Duffy gene covering the promoter region in the eight P. vivax-infected Cameroonians to identify the T-33C mutation revealed all these patients as Duffy-negative. The results provide evidence of single P. vivax as well as mixed malaria parasite infection in native Cameroonians and add knowledge to the growing evidences of P. vivax infection in Duffy-negative Africans.


Subject(s)
Duffy Blood-Group System , Malaria, Vivax/blood , Malaria, Vivax/parasitology , Malaria/blood , Malaria/parasitology , Plasmodium vivax/genetics , Base Sequence , Cameroon , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/genetics , Geography , Humans , Malaria/epidemiology , Malaria, Vivax/epidemiology , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Protozoan/genetics , RNA, Ribosomal, 18S/genetics , Sequence Alignment
7.
Transfusion ; 54(12): 3198-207, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24845979

ABSTRACT

BACKGROUND: After the ABO (ABO) and Rh (RHD and RHCE) blood group systems, Kell (KEL), Kidd (SLC14A1), and Duffy (DARC) represent the second most important clinically relevant antigens. STUDY DESIGN AND METHODS: Samples from 4000 Swiss blood donors, with serologic prevalues for K/k, Kp(a/b), Jk(a/b), and Fy(a/b), and 48 additional samples of presumptive black African origin were genotyped using high-throughput matrix-assisted laser desorption/ionization, time-of-flight mass spectrometry, applying one single-multiplex polymerase chain reaction/primer-extension reaction simultaneously detecting 15 single-nucleotide polymorphisms. RESULTS: Genotype/phenotype concordance for K/k, Kp(a/b), Jk(a/b), and all Fy(a/b) specificities were 100, 99.98, 99.93, and 99.20%, respectively. Discrepancies were caused by erroneous serologic profiles (n = 33), mainly attributed to weakly expressed Fy(x) (n = 28). Only three discrepancies had a genetic basis. They could all be explained by newly observed silenced alleles: one KEL*02N.34 and one FY*02N.03 with predicted R700Q and G261R amino acid exchanges, respectively, and one JK*B, with an as-yet-unidentified silencing cause. According to NCBI SNP database entry for rs8176034, another new allele, KEL*02.38, had been expected, and we formally demonstrated its presence. We furthermore identified individuals with rare phenotypes, such as Js(a/b) heterozygotes among Caucasians, rare alleles, the "Swiss" JK*01N.03, and rare genotypes, such as one Fy(x) homozygote. CONCLUSION: Genotyping proved its practicability in the daily routine setting and qualitatively outperformed serology. Technology is ideal for time-insensitive donor genotyping and allows for a broad range of throughput needs. Consequently, from a technologic point of view, serotyping should be replaced by genotyping for donors' blood groups encoded by KEL, SLC14A1, and DARC.


Subject(s)
Alleles , Duffy Blood-Group System/genetics , Genotyping Techniques/methods , Kidd Blood-Group System/genetics , Membrane Glycoproteins/genetics , Metalloendopeptidases/genetics , Receptors, Cell Surface/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adult , Databases, Nucleic Acid , Duffy Blood-Group System/chemistry , Female , Genotyping Techniques/instrumentation , Humans , Kidd Blood-Group System/chemistry , Male , Membrane Glycoproteins/chemistry , Metalloendopeptidases/chemistry , Middle Aged , Polymorphism, Single Nucleotide , Receptors, Cell Surface/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/instrumentation , Switzerland
8.
PLoS Pathog ; 10(1): e1003869, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24415938

ABSTRACT

Plasmodium parasites use specialized ligands which bind to red blood cell (RBC) receptors during invasion. Defining the mechanism of receptor recognition is essential for the design of interventions against malaria. Here, we present the structural basis for Duffy antigen (DARC) engagement by P. vivax Duffy binding protein (DBP). We used NMR to map the core region of the DARC ectodomain contacted by the receptor binding domain of DBP (DBP-RII) and solved two distinct crystal structures of DBP-RII bound to this core region of DARC. Isothermal titration calorimetry studies show these structures are part of a multi-step binding pathway, and individual point mutations of residues contacting DARC result in a complete loss of RBC binding by DBP-RII. Two DBP-RII molecules sandwich either one or two DARC ectodomains, creating distinct heterotrimeric and heterotetrameric architectures. The DARC N-terminus forms an amphipathic helix upon DBP-RII binding. The studies reveal a receptor binding pocket in DBP and critical contacts in DARC, reveal novel targets for intervention, and suggest that targeting the critical DARC binding sites will lead to potent disruption of RBC engagement as complex assembly is dependent on DARC binding. These results allow for models to examine inter-species infection barriers, Plasmodium immune evasion mechanisms, P. knowlesi receptor-ligand specificity, and mechanisms of naturally acquired P. vivax immunity. The step-wise binding model identifies a possible mechanism by which signaling pathways could be activated during invasion. It is anticipated that the structural basis of DBP host-cell engagement will enable development of rational therapeutics targeting this interaction.


Subject(s)
Antigens, Protozoan/chemistry , Duffy Blood-Group System/chemistry , Erythrocytes/chemistry , Plasmodium vivax/chemistry , Protozoan Proteins/chemistry , Receptors, Cell Surface/chemistry , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Cell Line , Duffy Blood-Group System/genetics , Duffy Blood-Group System/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Immune Evasion , Malaria, Vivax/genetics , Malaria, Vivax/immunology , Plasmodium vivax/immunology , Plasmodium vivax/metabolism , Point Mutation , Protein Binding , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Structure-Activity Relationship
9.
J Biol Chem ; 287(39): 32922-39, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22843685

ABSTRACT

Invasion of human red blood cells by Plasmodium falciparum involves interaction of the merozoite form through proteins on the surface coat. The erythrocyte binding-like protein family functions after initial merozoite interaction by binding via the Duffy binding-like (DBL) domain to receptors on the host red blood cell. The merozoite surface proteins DBL1 and -2 (PfMSPDBL1 and PfMSPDBL2) (PF10_0348 and PF10_0355) are extrinsically associated with the merozoite, and both have a DBL domain in each protein. We expressed and refolded recombinant DBL domains for PfMSPDBL1 and -2 and show they are functional. The red cell binding characteristics of these domains were shown to be similar to full-length forms of these proteins isolated from parasite cultures. Futhermore, metal cofactors were found to enhance the binding of both the DBL domains and the parasite-derived full-length proteins to erythrocytes, which has implications for receptor binding of other DBL-containing proteins in Plasmodium spp. We solved the structure of the erythrocyte-binding DBL domain of PfMSPDBL2 to 2.09 Å resolution and modeled that of PfMSPDBL1, revealing a canonical DBL fold consisting of a boomerang shaped α-helical core formed from three subdomains. PfMSPDBL2 is highly polymorphic, and mapping of these mutations shows they are on the surface, predominantly in the first two domains. For both PfMSPDBL proteins, polymorphic variation spares the cleft separating domains 1 and 2 from domain 3, and the groove between the two major helices of domain 3 extends beyond the cleft, indicating these regions are functionally important and are likely to be associated with the binding of a receptor on the red blood cell.


Subject(s)
Models, Molecular , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Crystallography, X-Ray , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/metabolism , Humans , Plasmodium falciparum/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Protozoan Proteins/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism
10.
Glycoconj J ; 29(2-3): 93-105, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22246380

ABSTRACT

Duffy antigen/receptor for chemokines (DARC) is a glycosylated seven-transmembrane protein acting as a blood group antigen, a chemokine binding protein and a receptor for Plasmodium vivax malaria parasite. It is present on erythrocytes and endothelial cells of postcapillary venules. The N-terminal extracellular domain of the Duffy glycoprotein carries Fy(a)/Fy(b) blood group antigens and Fy6 linear epitope recognized by monoclonal antibodies. Previously, we have shown that recombinant Duffy protein expressed in K562 cells has three N-linked oligosaccharide chains, which are mainly of complex-type. Here we report a one-step purification method of Duffy protein from human erythrocytes. DARC was extracted from erythrocyte membranes in the presence of 1% n-dodecyl-ß-D-maltoside (DDM) and 0.05% cholesteryl hemisuccinate (CHS) and purified by affinity chromatography using immobilized anti-Fy6 2C3 mouse monoclonal antibody. Duffy glycoprotein was eluted from the column with synthetic DFEDVWN peptide containing epitope for 2C3 monoclonal antibody. In this single-step immunoaffinity purification method we obtained highly purified DARC, which migrates in SDS-polyacrylamide gel as a major diffuse band corresponding to a molecular mass of 40-47 kDa. In ELISA purified Duffy glycoprotein binds anti-Duffy antibodies recognizing epitopes located on distinct regions of the molecule. Results of circular dichroism measurement indicate that purified DARC has a high content of α-helical secondary structure typical for chemokine receptors. Analysis of DARC glycans performed by means of lectin blotting and glycosidase digestion suggests that native Duffy N-glycans are mostly triantennary complex-type, terminated with α2-3- and α2-6-linked sialic acid residues with bisecting GlcNAc and α1-6-linked fucose at the core.


Subject(s)
Duffy Blood-Group System/isolation & purification , Erythrocyte Membrane/chemistry , Receptors, Cell Surface/isolation & purification , Receptors, Chemokine/isolation & purification , Antibodies, Monoclonal/chemistry , Chromatography, Affinity/methods , Circular Dichroism/methods , Duffy Blood-Group System/chemistry , Electrophoresis, Polyacrylamide Gel/methods , Glycoside Hydrolases/chemistry , Glycosylation , Humans , Lectins/chemistry , Peptide Fragments/chemistry , Polysaccharides/chemistry , Protein Binding , Protein Structure, Secondary , Receptors, Cell Surface/chemistry
11.
Nat Struct Mol Biol ; 18(8): 908-14, 2011 Jul 10.
Article in English | MEDLINE | ID: mdl-21743458

ABSTRACT

Plasmodium vivax and Plasmodium knowlesi invasion depends on the parasite Duffy-binding protein DBL domain (RII-PvDBP or RII-PkDBP) engaging the Duffy antigen receptor for chemokines (DARC) on red blood cells. Inhibition of this key interaction provides an excellent opportunity for parasite control. There are competing models for whether Plasmodium ligands engage receptors as monomers or dimers, a question whose resolution has profound implications for parasite biology and control. We report crystallographic, solution and functional studies of RII-PvDBP showing that dimerization is required for and driven by receptor engagement. This work provides a unifying framework for prior studies and accounts for the action of naturally acquired blocking antibodies and the mechanism of immune evasion. We show that dimerization is conserved in DBL-domain receptor engagement and propose that receptor-mediated ligand dimerization drives receptor affinity and specificity. Because dimerization is prevalent in signaling, our studies raise the possibility that induced dimerization may activate pathways for invasion.


Subject(s)
Antigens, Protozoan/metabolism , Duffy Blood-Group System/metabolism , Plasmodium vivax/metabolism , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Antigens, Protozoan/chemistry , Binding Sites , Crystallography, X-Ray , Dimerization , Duffy Blood-Group System/chemistry , Humans , Ligands , Models, Molecular , Plasmodium falciparum/metabolism , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Receptors, Cell Surface/chemistry , Signal Transduction
12.
Transfus Clin Biol ; 17(3): 184-96, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20655787

ABSTRACT

Duffy Antigen Receptor for Chemokines (DARC) is an unusual transmembrane chemokine receptor which (i) binds the two main chemokine families and (ii) does not transduct any signal as it lacks the DRY consensus sequence. It is considered as silent chemokine receptor, a tank useful for chemiotactism. DARC had been particularly studied as a major actor of malaria infection by Plasmodium vivax. It is also implicated in multiple chemokine inflammation, inflammatory diseases, in cancer and might play a role in HIV infection and AIDS. In this review, we focus on the interest to build structural model of DARC to understand more precisely its abilities to bind its physiological ligand CXCL8 and its malaria ligand. We also present innovative development on VHHs able to bind DARC protein. We underline difficulties and limitations of such bioinformatics approaches and highlight the crucial importance of biological data to conduct these kinds of researches.


Subject(s)
Duffy Blood-Group System/chemistry , Receptors, Cell Surface/chemistry , Amino Acid Sequence , Animals , Chemokines/metabolism , Chemotaxis , Computer Simulation , Consensus Sequence , Duffy Blood-Group System/genetics , Duffy Blood-Group System/metabolism , Humans , Malaria, Vivax/blood , Models, Molecular , Molecular Sequence Data , Mutation , Plasmodium vivax/pathogenicity , Plasmodium vivax/physiology , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid
13.
J Pept Sci ; 16(4): 190-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20196090

ABSTRACT

Tyrosine sulfation is a post translational modification that occurs on integral membrane and secreted proteins, and is required for mediating crucial biological processes. Until recently the synthesis of sTyr peptides, especially those containing multiple sTyr residues, were among the most challenging peptides to prepare. We recently described an efficient strategy for Fmoc-based solid phase synthesis of sTyr peptides in which the sulfate group in the sTyr residue(s) is protected with a DCV group (FmocTyr(SO(3)DCV)OH, 1). After cleavage of the peptide from the support the DCV group is removed by hydrogenolysis. Here we demonstrate that sTyr peptides containing Met or Trp residues can be prepared using our sulfate-protecting group strategy by preparing peptides corresponding to residues 1-20 of chemokine receptor CXCR6 and 8-42 of chemokine receptor DARC. Removing the DCV groups at the end of the syntheses was readily achieved, without any reduction of the indole ring in Trp, by performing the hydrogenolysis in the presence of triethylamine. These conditions were found to be particularly efficient for removing the DCV group and superiour to our original conditions using H(2), ammonium formate, Pd/C. The presence of Met was found not to interfere with the removal of the DCV group. The use of pseudoproline dipeptides and N-backbone protection with the 2,4-dimethoxybenzyl group were found to be very effective tactics for preventing aggregation and aspartimide formation during the synthesis of these peptides. We also report an alternative and more cost effective synthesis of amino acid 1.


Subject(s)
Duffy Blood-Group System/chemistry , Peptides/chemical synthesis , Receptors, Cell Surface/chemistry , Receptors, Chemokine/chemistry , Receptors, Virus/chemistry , Sulfates/chemistry , Animals , Chromatography, High Pressure Liquid , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Receptors, CXCR6 , Spectrometry, Mass, Electrospray Ionization
14.
Infect Disord Drug Targets ; 9(3): 289-303, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19519483

ABSTRACT

The Duffy Antigen/Receptor for Chemokine (DARC) is a seven segment transmembrane protein. It was firstly discovered as a blood group antigen and was the first specific gene locus assigned to a specific autosome in man. It became more famous as an erythrocyte receptor for malaria parasites (Plasmodium vivax and Plasmodium knowlesi), and finally for chemokines. DARC is an unorthodox chemokine receptor as (i) it binds chemokines of both CC and CXC classes and (ii) it lacks the Asp-Arg-Tyr consensus motif in its second cytoplasmic loop hence cannot couple to G proteins and activate their signaling pathways. DARC had also been associated to cancer progression, numerous inflammatory diseases, and possibly to AIDS. In this review, we will summarize important biological data on DARC. Then we shall focus on recent development of the elaboration and analyzes of structural models of DARC. We underline the difficulty to propose pertinent structural models of transmembrane protein using comparative modeling process, and other dedicated approaches as the Protein Blocks. The chosen structural models encompass most of the biochemical data known to date. Finally, we present recent development of protein-protein docking between DARC structural models and CXCL-8 structures. We propose a hierarchical search based on separated rigid and flexible docking.


Subject(s)
Duffy Blood-Group System/chemistry , Interleukin-8/metabolism , Receptors, Cell Surface/chemistry , Receptors, Chemokine/metabolism , Animals , Binding Sites , Computer Simulation , Duffy Blood-Group System/blood , Duffy Blood-Group System/genetics , Humans , Male , Models, Molecular , Plasmodium vivax/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Cell Surface/blood , Receptors, Cell Surface/genetics
15.
Methods Enzymol ; 461: 191-206, 2009.
Article in English | MEDLINE | ID: mdl-19480920

ABSTRACT

The Duffy blood group antigen is a serpentine protein with seven transmembrane domains that is not coupled to G-proteins or other known intracellular effectors. In addition to erythrocytes, it is also expressed in endothelial cells and neurons. In recent years the Duffy antigen has received much attention because of its diverse roles in health and disease. These include its functions as a docking receptor for the invasion of human erythrocytes by the malaria parasite Plasmodium vivax. In addition, the Duffy antigen is a binding protein for multiple inflammatory chemokines. Its expression allows erythrocytes to regulate intravascular levels of chemokines. It has also been shown recently that the Duffy antigen plays an important role in endothelial cells by facilitating chemokine transcytosis and presentation. Given these diverse functions of the Duffy antigen, this short review presents detailed methods that can be used to investigate each of these potential roles of this multifaceted protein.


Subject(s)
Chemokines/metabolism , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Chemokine/chemistry , Receptors, Chemokine/metabolism , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Humans , Models, Biological , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Cell Surface/genetics
16.
Mol Pharmacol ; 73(5): 1362-70, 2008 May.
Article in English | MEDLINE | ID: mdl-18230715

ABSTRACT

The Duffy antigen/receptor for chemokines (DARC) is an unusual chemokine receptor that binds a large number of inflammatory chemokines of both the CC and CXC families with nanomolar affinity, yet it lacks the ability to signal upon ligand binding. Using bioluminescent resonant energy transfer, we have demonstrated for the first time that DARC exists as a constitutive homo-oligomer in living cells and furthermore that DARC hetero-oligomerizes with the CC chemokine receptor CCR5. DARC-CCR5 interaction impairs chemotaxis and calcium flux through CCR5, whereas internalization of CCR5 in response to ligand binding remains unchanged. These results suggest a novel mechanism by which DARC could modulate inflammatory responses to chemokines in vivo.


Subject(s)
CCR5 Receptor Antagonists , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Arrestins/metabolism , Binding Sites , Calcium/metabolism , Cell Line , Cell Survival , Chemotaxis , Dimerization , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Ligands , Mice , Protein Binding , Protein Structure, Quaternary , Transfection , beta-Arrestins
17.
J Mol Biol ; 375(3): 773-81, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18036613

ABSTRACT

The malaria parasite proliferates in the bloodstream of its vertebrate host by invading and replicating within erythrocytes. To achieve successful invasion, a number of discrete and essential events need to take place at the parasite-host cell interface. Erythrocyte-binding antigen 175 (EBA-175) is a member of a family of Plasmodium falciparum erythrocyte-binding proteins involved in the formation of a tight junction, a necessary step in invasion. Here we present the crystal structure of EBA-175 region VI (rVI), a cysteine-rich domain that is highly conserved within the protein family and is essential for EBA-175 trafficking. The structure was solved by selenomethionine single-wavelength anomalous dispersion at 1.8 A resolution. It reveals a homodimer, containing in each subunit a compact five-alpha-helix core that is stabilized by four conserved disulfide bridges. rVI adopts a novel fold that is likely conserved across the protein family, indicating a conserved function. It shows no similarity to the Duffy-binding-like domains of EBA-175 involved in erythrocyte binding, indicating a distinct role. Remarkably, rVI possesses structural features related to the KIX-binding domain of the coactivator CREB-binding protein, supporting the binding and trafficking roles that have been ascribed to it and providing a rational basis for further experimental investigation of its function.


Subject(s)
Antigens, Protozoan , Duffy Blood-Group System/metabolism , Erythrocytes/metabolism , Malaria/blood , Plasmodium falciparum/chemistry , Plasmodium falciparum/metabolism , Protozoan Proteins/blood , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Cysteine/chemistry , Dimerization , Disulfides/chemistry , Duffy Blood-Group System/chemistry , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Molecular Weight , Plasmodium falciparum/pathogenicity , Protein Binding , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Water/chemistry
18.
Parasitol Res ; 101(4): 1045-50, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17533508

ABSTRACT

In areas where malaria transmission is stable, infants are often born to mothers who had Plasmodium falciparum infections during pregnancy. A significant number become exposed to infected erythrocytes or soluble parasite products with subsequent fetal immune priming or tolerance in utero. We performed ELISA to asses IgG and IgM seropositivity rates against three PfEMP1 DBL-alpha domains from 42 maternal-cord paired samples obtained at delivery from a hyperendemic area in Gabon. IgG was present in up to 80% of the cord serum samples, while IgM was found in only 20% of the same samples. These levels were not dependent on the parity of the mother or the peripheral and placental infectious status. The presence of IgM against DBL-alpha domain in cord serum samples suggests that this component is able to cross the placental barrier and mount a fetal immune response.


Subject(s)
Antibodies, Protozoan/blood , Duffy Blood-Group System/immunology , Fetal Blood/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Pregnancy Complications, Parasitic/immunology , Protozoan Proteins/immunology , Animals , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/genetics , Duffy Blood-Group System/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fetal Blood/parasitology , Gabon , Glutathione Transferase/genetics , Glutathione Transferase/immunology , Glutathione Transferase/metabolism , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Maternal-Fetal Exchange , Placenta , Pregnancy , Pregnancy Complications, Parasitic/diagnosis , Pregnancy Complications, Parasitic/parasitology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism
19.
Biochem Biophys Res Commun ; 356(3): 816-21, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17382291

ABSTRACT

The Duffy antigen/receptor for chemokines (DARC) is a seven-transmembrane glycoprotein carrying the Duffy (Fy) blood group antigen. The polypeptide chain of DARC contains two NSS motifs at positions 16 and 27 and one NDS motif at position 33 that represent canonical sequences for efficient N-glycosylation. To verify whether all of these three sites are occupied by a sugar chain, we generated mutants in which potential N-glycosylation sites (AsnXSer) were removed by replacement of serine by alanine. Seven DARC glycosylation variants, missing one (S18A, S29A, S35A), two (S18A.S29A, S18A.S35A, S29A.S35A), or three (S18A.S29A.S35A) glycosylation sites, were obtained. cDNA encoding DARC mutants was cloned into the eukaryotic expression vector pcDNA3.1/myc-HisA and expressed in human K562 cells. Stable transfectants expressing wild-type or mutated forms of Duffy were then lysed, purified by metal-affinity chromatography, and subjected to Western blots with an anti-Duffy monoclonal antibody. The gel electrophoresis data indicate that all three canonical sites are used for sugar attachment.


Subject(s)
Duffy Blood-Group System/genetics , Receptors, Cell Surface/genetics , Amino Acid Sequence , Amino Acid Substitution , Antibodies, Monoclonal , Blotting, Western , Chromatography, Affinity , DNA Mutational Analysis , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/immunology , Electrophoresis, Polyacrylamide Gel , Glycosylation , Humans , K562 Cells , Molecular Sequence Data , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology
SELECTION OF CITATIONS
SEARCH DETAIL