Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 340
Filter
1.
Protein Sci ; 33(6): e4999, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723106

ABSTRACT

Ticks produce chemokine-binding proteins, known as evasins, in their saliva to subvert the host's immune response. Evasins bind to chemokines and thereby inhibit the activation of their cognate chemokine receptors, thus suppressing leukocyte recruitment and inflammation. We recently described subclass A3 evasins, which, like other class A evasins, exclusively target CC chemokines but appear to use a different binding site architecture to control target selectivity among CC chemokines. We now describe the structural basis of chemokine recognition by the class A3 evasin EVA-ACA1001. EVA-ACA1001 binds to almost all human CC chemokines and inhibits receptor activation. Truncation mutants of EVA-ACA1001 showed that, unlike class A1 evasins, both the N- and C-termini of EVA-ACA1001 play minimal roles in chemokine binding. To understand the structural basis of its broad chemokine recognition, we determined the crystal structure of EVA-ACA1001 in complex with the human chemokine CCL16. EVA-ACA1001 forms backbone-backbone interactions with the CC motif of CCL16, a conserved feature of all class A evasin-chemokine complexes. A hydrophobic pocket in EVA-ACA1001, formed by several aromatic side chains and the unique disulfide bond of class A3 evasins, accommodates the residue immediately following the CC motif (the "CC + 1 residue") of CCL16. This interaction is shared with EVA-AAM1001, the only other class A3 evasins characterized to date, suggesting it may represent a common mechanism that accounts for the broad recognition of CC chemokines by class A3 evasins.


Subject(s)
Models, Molecular , Humans , Animals , Ticks/chemistry , Ticks/metabolism , Crystallography, X-Ray , Binding Sites , Arthropod Proteins/chemistry , Arthropod Proteins/metabolism , Arthropod Proteins/genetics , Protein Binding , Chemokines/chemistry , Chemokines/metabolism , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/metabolism
2.
Biopolymers ; 115(2): e23557, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37341434

ABSTRACT

Chemokines are important immune system proteins, many of which mediate inflammation due to their function to activate and cause chemotaxis of leukocytes. An important anti-inflammatory strategy is therefore to bind and inhibit chemokines, which leads to the need for biophysical studies of chemokines as they bind various possible partners. Because a successful anti-chemokine drug should bind at low concentrations, techniques such as fluorescence anisotropy that can provide nanomolar signal detection are required. To allow fluorescence experiments to be carried out on chemokines, a method is described for the production of fluorescently labeled chemokines. First, a fusion-tagged chemokine is produced in Escherichia coli, then efficient cleavage of the N-terminal fusion partner is carried out with lab-produced enterokinase, followed by covalent modification with a fluorophore, mediated by the lab-produced sortase enzyme. This overall process reduces the need for expensive commercial enzymatic reagents. Finally, we utilize the product, vMIP-fluor, in binding studies with the chemokine binding protein vCCI, which has great potential as an anti-inflammatory therapeutic, showing a binding constant for vCCI:vMIP-fluor of 0.37 ± 0.006 nM. We also show how a single modified chemokine homolog (vMIP-fluor) can be used in competition assays with other chemokines and we report a Kd for vCCI:CCL17 of 14 µM. This work demonstrates an efficient method of production and fluorescent labeling of chemokines for study across a broad range of concentrations.


Subject(s)
Chemokines, CC , Enteropeptidase , Humans , Chemokines, CC/chemistry , Chemokines, CC/metabolism , Chemokines/chemistry , Chemokines/metabolism , Inflammation , Anti-Inflammatory Agents
3.
Biochemistry ; 62(17): 2530-2540, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37540799

ABSTRACT

We investigate the physicochemical effects of pyroglutamination on the QHALTSV-NH2 peptide, a segment of cytosolic helix 8 of the human C-X-C chemokine G-protein-coupled receptor type 4 (CXCR4). This modification, resulting from the spontaneous conversion of glutamine to pyroglutamic acid, has significant impacts on the physicochemical features of peptides. Using a static approach, we compared the transformation in different conditions and experimentally found that the rate of product formation increases with temperature, underscoring the need for caution during laboratory experiments to prevent glutamine cyclization. Circular dichroism experiments revealed that the QHALTSV-NH2 segment plays a minor role in the structuration of H8 CXCR4; however, its pyroglutaminated analogue interacts differently with its chemical environment, showing increased susceptibility to solvent variations compared to the native form. The pyroglutaminated analogue exhibits altered behavior when interacting with lipid models, suggesting a significant impact on its interaction with cell membranes. A unique combination of atomic force microscopy and infrared nanospectroscopy revealed that pyroglutamination affects supramolecular self-assembly, leading to highly packed molecular arrangements and a crystalline structure. Moreover, the presence of pyroglumatic acid has been found to favor the formation of amyloidogenic aggregates. Our findings emphasize the importance of considering pyroglutamination in peptide synthesis and proteomics and its potential significance in amyloidosis.


Subject(s)
Amyloidosis , Glutamine , Humans , Peptides , Chemokines/chemistry , Cell Membrane/metabolism , Circular Dichroism , Receptors, CXCR4/metabolism
4.
J Med Chem ; 66(11): 7070-7085, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37212620

ABSTRACT

The chemokine system is a key player in the functioning of the immune system and a sought-after target for drug candidates. The number of experimental structures of chemokines in complex with chemokine receptors has increased rapidly over the past few years, providing essential information for rational development of chemokine receptor ligands. Here, we perform a comparative analysis of all chemokine-chemokine receptor structures, with the aim of characterizing the molecular recognition processes and highlighting the relationships between chemokine structures and functional processes. The structures show conserved interaction patterns between the chemokine core and the receptor N-terminus, while interactions near ECL2 display subfamily-specific features. Detailed analyses of the interactions of the chemokine N-terminal domain in the 7TM cavities reveal activation mechanisms for CCR5, CCR2, and CXCR2 and a mechanism for biased agonism in CCR1.


Subject(s)
Chemokines , Receptors, Chemokine , Chemokines/chemistry , Protein Binding , Receptors, CCR5/metabolism , Receptors, CCR1/metabolism
5.
Histopathology ; 82(3): 495-503, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36345263

ABSTRACT

AIMS: Classic Hodgkin lymphoma (cHL) should be distinguished from its wide variety of histological mimics, including reactive conditions and mature B and T cell neoplasms. Thymus and activation-related chemokine (TARC) is produced in extremely high quantities by the Hodgkin/Reed-Sternberg (HRS) tumour cells and is largely responsible for the attraction of CD4+ T cells into the cHL tumour micro-environment. In the current study we evaluated the diagnostic potential of TARC immunohistochemistry in daily practice in a tertiary referral centre in the Netherlands. METHODS AND RESULTS: A total of 383 cases, approximately half of which were cHL mimics, were prospectively evaluated in the period from June 2014 to November 2020. In 190 cHL cases, 92% were TARC-positive and the majority of cases showed strong and highly specific staining in all HRS cells (77%). In most cases, TARC could discriminate between nodular lymphocyte-predominant and lymphocyte-rich Hodgkin lymphoma. HRS-like cells in mature lymphoid neoplasms were rarely positive (6.4%) and there was no TARC staining at all in 64 reactive lymphadenopathies. CONCLUSIONS: TARC immunohistochemistry has great value in differentiating between cHL and its mimics, including nodular lymphocyte-predominant Hodgkin lymphoma, reactive lymphadenopathies and mature lymphoid neoplasms with HRS-like cells.


Subject(s)
Hodgkin Disease , Lymphadenopathy , Thymus Gland , Humans , Chemokines/chemistry , Chemokines/immunology , Hodgkin Disease/diagnosis , Hodgkin Disease/pathology , Immunohistochemistry , Lymphadenopathy/pathology , Reed-Sternberg Cells/pathology , Tumor Microenvironment , Thymus Gland/immunology , Thymus Gland/metabolism
6.
J Struct Biol ; 214(3): 107877, 2022 09.
Article in English | MEDLINE | ID: mdl-35750237

ABSTRACT

Chemokine receptors are the central signaling hubs of several processes such as cell migration, chemotaxis and cell positioning. In this graphical review, we provide an overview of the structural and mechanistic principles governing chemokine recognition that are currently emerging. Structural models of chemokine-receptor co-complexes with endogenous chemokines, viral chemokines and therapeutics have been resolved that highlight multiple interaction sites, termed as CRS1, CRS1.5 etc. The first site of interaction has been shown to be the N-terminal domain of the receptors (CRS1 site). A large structural flexibility of the N-terminal domain has been reported that was supported by both experimental and simulation studies. Upon chemokine binding, the N-terminal domain appears to show constricted dynamics and opens up to interact with the chemokine via a large interface. The subsequent sites such as CRS1.5 and CRS2 sites have been structurally well resolved although differences arise such as the localization of the N-terminus of the ligand to a major or minor pocket of the orthosteric binding site. Several computational studies have highlighted the dynamic protein-protein interface at the CRS1 site that seemingly appears to resolve the differences in NMR and mutagenesis studies. Interestingly, the differential dynamics at the CRS1 site suggests a mixed model of binding with complex signatures of both conformational selection and induced fit models. Integrative experimental and computational approaches could help unravel the structural basis of promiscuity and specificity in chemokine-receptor binding and open up new avenues of therapeutic design.


Subject(s)
Receptors, Chemokine , Signal Transduction , Binding Sites , Chemokines/chemistry , Chemokines/metabolism , Protein Binding , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
7.
Chem Commun (Camb) ; 58(26): 4132-4148, 2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35274633

ABSTRACT

Ever since the first biologically active chemokines were discovered in the late 1980s, these messenger proteins and their receptors have been the target for a plethora of drug discovery efforts in the pharmaceutical industry, as well as in academia. Owing to the publication of several chemokine receptor X-ray crystal structures, a highly druggable, intracellular, allosteric binding site which partially overlaps with the G protein binding site was discovered. This intriguing, new approach for chemokine receptor antagonism has captured researchers around the world, pushing the exploration of this intracellular binding site and new antagonists thereof. In this review, we have highlighted the past two decades of research on small-molecule chemokine receptor antagonists that modulate receptor function at the intracellular binding site.


Subject(s)
Chemokines , Drug Discovery , Allosteric Site , Binding Sites , Chemokines/chemistry , Chemokines/metabolism
8.
J Med Chem ; 64(20): 15349-15366, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34662112

ABSTRACT

To design novel antimicrobial peptides by utilizing the sequence of the human host defense protein, chemerin, a seven-residue amphipathic stretch located in the amino acid region, 109-115, was identified, which possesses the highest density of hydrophobic and positively charged residues. Although this 7-mer peptide was inactive toward microorganisms, its 14-mer tandem repeat (Chem-KVL) was highly active against different bacteria including methicillin-resistant Staphylococcus aureus, a multidrug-resistant Staphylococcus aureus strain, and slow- and fast-growing mycobacterial species. The selective enantiomeric substitutions of its two l-lysine residues were attempted to confer cell selectivity and proteolytic stability to Chem-KVL. Chem-8dK with a d-lysine replacement in its middle (eighth position) showed the lowest hemolytic activity against human red blood cells among Chem-KVL analogues and maintained high antimicrobial properties. Chem-8dK showed in vivo efficacy against Pseudomonas aeruginosa infection in BALB/c mice and inhibited the development of resistance in this microorganism up to 30 serial passages and growth of intracellular mycobacteria in THP-1 cells.


Subject(s)
Antimicrobial Peptides/pharmacology , Antitubercular Agents/pharmacology , Chemokines/chemistry , Lysine/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Pseudomonas aeruginosa/drug effects , Animals , Antimicrobial Peptides/chemical synthesis , Antimicrobial Peptides/chemistry , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Molecular Structure , Pseudomonas Infections/drug therapy , Stereoisomerism , Structure-Activity Relationship , THP-1 Cells
10.
Cell Mol Life Sci ; 78(17-18): 6265-6281, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34241650

ABSTRACT

Tight regulation of cytokines is essential for the initiation and resolution of inflammation. Chemerin, a mediator of innate immunity, mainly acts on chemokine-like receptor 1 (CMKLR1) to induce the migration of macrophages and dendritic cells. The role of the second chemerin receptor, G protein-coupled receptor 1 (GPR1), is still unclear. Here we demonstrate that GPR1 shows ligand-induced arrestin3 recruitment and internalization. The chemerin C-terminus triggers this activation by folding into a loop structure, binding to aromatic residues in the extracellular loops of GPR1. While this overall binding mode is shared between GPR1 and CMKLR1, differences in their respective extracellular loop 2 allowed for the design of the first GPR1-selective peptide. However, our results suggest that ligand-induced arrestin recruitment is not the only mode of action of GPR1. This receptor also displays constitutive internalization, which allows GPR1 to internalize inactive peptides efficiently by an activation-independent pathway. Our results demonstrate that GPR1 takes a dual role in regulating chemerin activity: as a signaling receptor for arrestin-based signaling on one hand, and as a scavenging receptor with broader ligand specificity on the other.


Subject(s)
Ligands , Receptors, G-Protein-Coupled/metabolism , Arrestins/metabolism , Binding Sites , Chemokines/chemistry , Chemokines/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Immunity, Innate , Microscopy, Confocal , Molecular Docking Simulation , Mutagenesis , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Chemokine/chemistry , Receptors, Chemokine/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics
11.
Nat Commun ; 12(1): 4151, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34230484

ABSTRACT

The chemokine receptor CCR5 plays a vital role in immune surveillance and inflammation. However, molecular details that govern its endogenous chemokine recognition and receptor activation remain elusive. Here we report three cryo-electron microscopy structures of Gi1 protein-coupled CCR5 in a ligand-free state and in complex with the chemokine MIP-1α or RANTES, as well as the crystal structure of MIP-1α-bound CCR5. These structures reveal distinct binding modes of the two chemokines and a specific accommodate pattern of the chemokine for the distal N terminus of CCR5. Together with functional data, the structures demonstrate that chemokine-induced rearrangement of toggle switch and plasticity of the receptor extracellular region are critical for receptor activation, while a conserved tryptophan residue in helix II acts as a trigger of receptor constitutive activation.


Subject(s)
Chemokines/chemistry , Chemokines/metabolism , Receptors, CCR5/chemistry , Receptors, CCR5/metabolism , Binding Sites , Chemokine CCL3/metabolism , Chemokine CCL5/chemistry , Chemokine CCL5/metabolism , Cryoelectron Microscopy , Ligands , Models, Molecular , Protein Conformation , Receptors, CCR5/genetics
12.
J Med Chem ; 64(6): 3048-3058, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33705662

ABSTRACT

The chemokine-like receptor 1 (CMKLR1) is a promising target for treating autoinflammatory diseases, cancer, and reproductive disorders. However, the interaction between CMKLR1 and its protein-ligand chemerin remains uncharacterized, and no drugs targeting this interaction have passed clinical trials. Here, we identify the binding mode of chemerin-9, the C-terminus of chemerin, at the receptor by combining complementary mutagenesis with structure-based modeling. Incorporating our experimental data, we present a detailed model of this binding site, including experimentally confirmed pairwise interactions for the most critical ligand residues: Chemerin-9 residue F8 binds to a hydrophobic pocket in CMKLR1 formed by the extracellular loop (ECL) 2, while F6 interacts with Y2.68, suggesting a turn-like structure. On the basis of this model, we created the first cyclic peptide with nanomolar activity, confirming the overall binding conformation. This constrained agonist mimics the loop conformation adopted by the natural ligand and can serve as a lead compound for future drug design.


Subject(s)
Chemokines/chemistry , Chemokines/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Receptors, Chemokine/agonists , Animals , Binding Sites , Cattle , Drug Discovery , Humans , Mice , Molecular Docking Simulation , Protein Conformation , Receptors, Chemokine/metabolism
14.
Front Immunol ; 11: 1629, 2020.
Article in English | MEDLINE | ID: mdl-32849553

ABSTRACT

Pathological self-assembly is a concept that is classically associated with amyloids, such as amyloid-ß (Aß) in Alzheimer's disease and α-synuclein in Parkinson's disease. In prokaryotic organisms, amyloids are assembled extracellularly in a similar fashion to human amyloids. Pathogenicity of amyloids is attributed to their ability to transform into several distinct structural states that reflect their downstream biological consequences. While the oligomeric forms of amyloids are thought to be responsible for their cytotoxicity via membrane permeation, their fibrillar conformations are known to interact with the innate immune system to induce inflammation. Furthermore, both eukaryotic and prokaryotic amyloids can self-assemble into molecular chaperones to bind nucleic acids, enabling amplification of Toll-like receptor (TLR) signaling. Recent work has shown that antimicrobial peptides (AMPs) follow a strikingly similar paradigm. Previously, AMPs were thought of as peptides with the primary function of permeating microbial membranes. Consistent with this, many AMPs are facially amphiphilic and can facilitate membrane remodeling processes such as pore formation and fusion. We show that various AMPs and chemokines can also chaperone and organize immune ligands into amyloid-like ordered supramolecular structures that are geometrically optimized for binding to TLRs, thereby amplifying immune signaling. The ability of amphiphilic AMPs to self-assemble cooperatively into superhelical protofibrils that form structural scaffolds for the ordered presentation of immune ligands like DNA and dsRNA is central to inflammation. It is interesting to explore the notion that the assembly of AMP protofibrils may be analogous to that of amyloid aggregates. Coming full circle, recent work has suggested that Aß and other amyloids also have AMP-like antimicrobial functions. The emerging perspective is one in which assembly affords a more finely calibrated system of recognition and response: the detection of single immune ligands, immune ligands bound to AMPs, and immune ligands spatially organized to varying degrees by AMPs, result in different immunologic outcomes. In this framework, not all ordered structures generated during multi-stepped AMP (or amyloid) assembly are pathological in origin. Supramolecular structures formed during this process serve as signatures to the innate immune system to orchestrate immune amplification in a proportional, situation-dependent manner.


Subject(s)
Amyloid/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Amyloid/chemistry , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/metabolism , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmunity , Biomarkers , Chemokines/chemistry , Chemokines/pharmacology , Host-Pathogen Interactions/immunology , Humans , Immunity , Immunity, Innate , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Ligands , Molecular Dynamics Simulation , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Pore Forming Cytotoxic Proteins/chemistry , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Protein Conformation , Structure-Activity Relationship , Toll-Like Receptors/chemistry , Toll-Like Receptors/metabolism
15.
Handb Exp Pharmacol ; 262: 231-258, 2020.
Article in English | MEDLINE | ID: mdl-32661663

ABSTRACT

Chemokines are a family of small proteins, subdivided by their conserved cysteine residues and common structural features. Chemokines interact with their cognate G-protein-coupled receptors to elicit downstream signals that result in cell migration, proliferation, and survival. This review presents evidence for how the various CXC and CC subfamily chemokines influence bone hemostasis by acting on osteoclasts, osteoblasts, and progenitor cells. Also discussed are the ways in which chemokines contribute to bone loss as a result of inflammatory diseases such as rheumatoid arthritis, HIV infection, and periodontal infection. Both positive and negative effects of chemokines on bone formation and bone loss are presented. In addition, the role of chemokines in altering the bone microenvironment through effects on angiogenesis and tumor invasion is discussed. Very few therapeutic agents that influence bone formation by targeting chemokines or chemokine receptors are available, although a few are currently being evaluated.


Subject(s)
Chemokines/chemistry , HIV Infections , Bone and Bones , Chemokines/immunology , Humans , Osteoblasts/chemistry , Osteoblasts/physiology , Osteoclasts/chemistry , Osteoclasts/physiology
16.
Exp Biol Med (Maywood) ; 245(16): 1518-1528, 2020 10.
Article in English | MEDLINE | ID: mdl-32715782

ABSTRACT

IMPACT STATEMENT: CKLF1, a recently identified chemokine, has been reported by a number of studies to play important roles in quite many diseases. However, the potential pathways that CKLF1 may be involved are not manifested well yet. In our review, we showed the basic molecular structure and major functions of this novel chemokine, and implication in human diseases, such as tumors. To attract more attention, we summarized its signaling pathways and clearly present them in a set of figures. With the overview of the experimental trial of CKLF1-targeting medicines in animal models, we hope to provide a few important insights about CKLF1 to both medical researchers and pharmacy.


Subject(s)
Chemokines/metabolism , Disease , Molecular Targeted Therapy , Animals , Chemokines/chemistry , Chemokines/genetics , Chemotaxis , Humans , Models, Biological , Signal Transduction
17.
Int J Biol Macromol ; 156: 239-251, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32289428

ABSTRACT

Chemokines are a sub-group of cytokines that regulate the leukocyte migration. Monocyte chemoattractant protein-1 (MCP/CCL2) is one of the essential CC chemokine that regulates the migration of monocytes into inflamed tissues. It has been observed that the primary sequences of CCL2 orthologs among rodents and primates vary significantly at the C-terminal region. However, no structural details are available for the rodentia family CCL2 proteins. The current study unravelled the structural, dynamics and in-silico functional characteristics of murine CCL2 chemokine using a comprehensive set of NMR spectroscopy techniques and evolutionary approaches. The study unravelled that the N-terminal portion of the murine CCL2 forms a canonical CC chemokine dimer similar to that of human CCL2. However, unlike human CCL2, the murine ortholog exhibits extensive dynamics in the µs-ms timescales. The presence of C-terminal region of the murine CCL2 protein/rodentia family is highly glycosylated, completely disordered, and inhibits the folding of the structured CCL2 regions. Further, it has been observed that the glycosaminoglycan binding surfaces of these orthologs proteins are greatly differed. In a nut shell, this comparative study provided the role of molecular evolution in generating orthologous proteins with differential structural and dynamics characteristics to engage them in specific molecular interactions.


Subject(s)
Chemokine CCL2/chemistry , Models, Molecular , Protein Conformation , Amino Acid Sequence , Animals , Binding Sites , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokines/chemistry , Chemokines/metabolism , Cloning, Molecular , Evolution, Molecular , Gene Expression , Magnetic Resonance Spectroscopy , Mice , Phylogeny , Protein Binding , Protein Multimerization , Recombinant Proteins , Spectrum Analysis , Structure-Activity Relationship
18.
Biochem Biophys Res Commun ; 528(2): 347-358, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32145914

ABSTRACT

The chemokines receptor family are membrane-expressed class A-specific seven-transmembrane receptors linked to G proteins. Through interaction with the corresponding ligands, the chemokines, they induce a wide variety of cellular responses including cell polarization, movement, immune and inflammatory responses, as well as the prevention of HIV-1 infection. Like a Russian matryoshka doll, the chemokine receptor system is more complex than initially envisaged. This review focuses on the mechanisms that contribute to this dazzling complexity and how they modulate the signaling events triggered by chemokines. The chemokines and their receptors exist as monomers, dimers and oligomers, their expression pattern is highly regulated, and the ligands can bind distinct receptors with similar affinities. The use of novel imaging-based technologies, particularly real-time imaging modalities, has shed new light on the very dynamic conformations that chemokine receptors adopt depending on the cellular context, and that affect chemokine-mediated responses. This complex scenario presents both challenging and exciting opportunities for drug discovery.


Subject(s)
Receptors, Chemokine/metabolism , Animals , Chemokines/chemistry , Chemokines/metabolism , Chemotactic Factors/metabolism , Humans , Protein Multimerization
19.
Article in English | MEDLINE | ID: mdl-31997766

ABSTRACT

Inflammation, is driven by a network comprising cytokines, chemokines, their target receptors and leukocytes, and is a major pathologic mechanism that adversely affects organ function in diverse human diseases. Despite being supported by substantial target validation, no successful anti-chemokine therapeutic to treat inflammatory disease has yet been developed. This is in part because of the robustness of the chemokine network, which emerges from a large total chemokine load in disease, promiscuous expression of receptors on leukocytes, promiscuous and synergistic interactions between chemokines and receptors, and feedforward loops created by secretion of chemokines by leukocytes themselves. Many parasites, including viruses, helminths and ticks, evade the chemokine network by producing proteins that bind promiscuously to chemokines or their receptors. Evasins - three small glycoproteins identified in the saliva of the brown dog tick - bind multiple chemokines, and are active in several animal models of inflammatory disease. Over 50 evasin homologs have recently been identified from diverse tick species. Characterization of the chemokine binding patterns of evasins show that several have anti-chemokine activities that extend substantially beyond those previously described. These studies indicate that evasins function at the site of the tick bite by reducing total chemokine load. This not only reduces chemokine signaling to receptors, but also interrupts feedforward loops, thus disabling the chemokine network. Taking the lead from nature, a goal for the development of new anti-chemokine therapeutics would be to reduce the total chemokine load in disease. This could be achieved by administering appropriate evasin combinations or by smaller peptides that mimic evasin action.


Subject(s)
Chemokines/metabolism , Inflammation/metabolism , Insect Proteins/metabolism , Animals , Chemokines/chemistry , Humans , Peptidomimetics
20.
J Leukoc Biol ; 107(6): 1115-1122, 2020 06.
Article in English | MEDLINE | ID: mdl-31965639

ABSTRACT

Chemokines are small soluble proteins that drive cell migration through the formation of concentration gradients. Chemokine binding to G protein-coupled chemokine receptors in the cell membrane activates intracellular signaling pathways and is a fundamental process involved in numerous physiological and pathophysiological functions. In the past few years, significant experimental developments have made it possible to characterize complexes between chemokine receptors and chemokines at a molecular level. Here, I review these developments from an experimental perspective, focusing on how the ability to express, purify, and stabilize receptor:chemokine complexes have made studies by X-ray crystallography, nuclear magnetic resonance, and other methods possible. I give examples of how these studies have advanced our understanding of the architecture of receptor:chemokine complexes as well as the mechanisms involved in complex formation. Finally, I discuss some of the many remaining questions and challenges that will require studies of more receptors and chemokines as well as further development of experimental methods.


Subject(s)
Chemokines/chemistry , Glycosaminoglycans/chemistry , Receptors, Chemokine/chemistry , Binding Sites , Chemokines/genetics , Chemokines/metabolism , Crystallography, X-Ray/methods , Gene Expression , Glycosaminoglycans/metabolism , HEK293 Cells , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...