Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 344
Filtrar
1.
J Am Soc Mass Spectrom ; 35(7): 1550-1555, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38836362

RESUMEN

Heparin, a widely used clinical anticoagulant, is generally well-tolerated; however, approximately 1% of patients develop heparin-induced thrombocytopenia (HIT), a serious side effect. While efforts to understand the role of chemokines in HIT development are ongoing, certain aspects remain less studied, such as the stabilization of chemokine oligomers by heparin. Here, we conducted a combined ion mobility-native mass spectrometry study to investigate the stability of chemokine oligomers and their complexes with fondaparinux, a synthetic heparin analog. Collision-induced dissociation and unfolding experiments provided clarity on the specificity and relevance of chemokine oligomers and their fondaparinux complexes with varying stoichiometries, as well as the stabilizing effects of fondaparinux binding.


Asunto(s)
Anticoagulantes , Fondaparinux , Polisacáridos , Fondaparinux/química , Fondaparinux/farmacología , Polisacáridos/química , Polisacáridos/metabolismo , Anticoagulantes/química , Anticoagulantes/farmacología , Quimiocinas/química , Quimiocinas/metabolismo , Humanos , Heparina/química , Heparina/metabolismo , Unión Proteica , Espectrometría de Movilidad Iónica/métodos , Espectrometría de Masas/métodos
2.
Biochem Soc Trans ; 52(3): 1011-1024, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38856028

RESUMEN

Chemokine receptors are integral to the immune system and prime targets in drug discovery that have undergone extensive structural elucidation in recent years. We outline a timeline of these structural achievements, discuss the intracellular negative allosteric modulation of chemokine receptors, analyze the mechanisms of orthosteric receptor activation, and report on the emerging concept of biased signaling. Additionally, we highlight differences of G-protein binding among chemokine receptors. Intracellular allosteric modulators in chemokine receptors interact with a conserved motif within transmembrane helix 7 and helix 8 and exhibit a two-fold inactivation mechanism that can be harnessed for drug-discovery efforts. Chemokine recognition is a multi-step process traditionally explained by a two-site model within chemokine recognition site 1 (CRS1) and CRS2. Recent structural studies have extended our understanding of this complex mechanism with the identification of CRS1.5 and CRS3. CRS3 is implicated in determining ligand specificity and surrounds the chemokine by almost 180°. Within CRS3 we identified the extracellular loop 2 residue 45.51 as a key interaction mediator for chemokine binding. Y2917.43 on the other hand was shown in CCR1 to be a key determinant of signaling bias which, along with specific chemokine-dependent phosphorylation ensembles at the G-protein coupled receptors (GPCR's) C-terminus, seems to play a pivotal role in determining the direction of signal bias in GPCRs.


Asunto(s)
Receptores de Quimiocina , Transducción de Señal , Receptores de Quimiocina/metabolismo , Receptores de Quimiocina/química , Humanos , Quimiocinas/metabolismo , Quimiocinas/química , Unión Proteica , Regulación Alostérica , Modelos Moleculares , Animales , Sitios de Unión , Conformación Proteica , Ligandos
3.
ACS Chem Biol ; 19(7): 1426-1432, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-38941516

RESUMEN

Chemokines are an important family of small proteins integral to leukocyte recruitment during inflammation. Dysregulation of the chemokine-chemokine receptor axis is implicated in many diseases, and both chemokines and their cognate receptors have been the targets of therapeutic development. Analysis of the antigen-binding regions of chemokine-binding nanobodies revealed a sequence motif suggestive of tyrosine sulfation. Given the well-established importance of post-translational tyrosine sulfation of receptors for chemokine affinity, it was hypothesized that the sulfation of these nanobodies may contribute to chemokine binding and selectivity. Four nanobodies (16C1, 9F1, 11B1, and 11F2) were expressed using amber codon suppression to incorporate tyrosine sulfation. The sulfated variant of 16C1 demonstrated significantly improved chemokine binding compared to the non-sulfated counterpart, while the other nanobodies displayed equipotent or reduced affinity upon sulfation. The ability of tyrosine sulfation to modulate chemokine binding, both positively and negatively, could be leveraged for chemokine-targeted sulfo-nanobody therapeutics in the future.


Asunto(s)
Quimiocinas , Anticuerpos de Dominio Único , Tirosina , Tirosina/metabolismo , Tirosina/química , Tirosina/análogos & derivados , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo , Quimiocinas/metabolismo , Quimiocinas/química , Humanos , Unión Proteica , Sulfatos/metabolismo , Sulfatos/química
4.
J Chem Inf Model ; 64(11): 4587-4600, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38809680

RESUMEN

AlphaFold and AlphaFold-Multimer have become two essential tools for the modeling of unknown structures of proteins and protein complexes. In this work, we extensively benchmarked the quality of chemokine-chemokine receptor structures generated by AlphaFold-Multimer against experimentally determined structures. Our analysis considered both the global quality of the model, as well as key structural features for chemokine recognition. To study the effects of template and multiple sequence alignment parameters on the results, a new prediction pipeline called LIT-AlphaFold (https://github.com/LIT-CCM-lab/LIT-AlphaFold) was developed, allowing extensive input customization. AlphaFold-Multimer correctly predicted differences in chemokine binding orientation and accurately reproduced the unique binding orientation of the CXCL12-ACKR3 complex. Further, the predictions of the full receptor N-terminus provided insights into a putative chemokine recognition site 0.5. The accuracy of chemokine N-terminus binding mode prediction varied between complexes, but the confidence score permitted the distinguishing of residues that were very likely well positioned. Finally, we generated a high-confidence model of the unsolved CXCL12-CXCR4 complex, which agreed with experimental mutagenesis and cross-linking data.


Asunto(s)
Benchmarking , Quimiocinas , Modelos Moleculares , Conformación Proteica , Quimiocinas/metabolismo , Quimiocinas/química , Receptores de Quimiocina/metabolismo , Receptores de Quimiocina/química , Unión Proteica , Humanos , Secuencia de Aminoácidos
5.
Protein Sci ; 33(6): e4999, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38723106

RESUMEN

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.


Asunto(s)
Modelos Moleculares , Humanos , Animales , Garrapatas/química , Garrapatas/metabolismo , Cristalografía por Rayos X , Sitios de Unión , Proteínas de Artrópodos/química , Proteínas de Artrópodos/metabolismo , Proteínas de Artrópodos/genética , Unión Proteica , Quimiocinas/química , Quimiocinas/metabolismo , Proteínas y Péptidos Salivales/química , Proteínas y Péptidos Salivales/metabolismo
6.
Biopolymers ; 115(2): e23557, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37341434

RESUMEN

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.


Asunto(s)
Quimiocinas CC , Enteropeptidasa , Humanos , Quimiocinas CC/química , Quimiocinas CC/metabolismo , Quimiocinas/química , Quimiocinas/metabolismo , Inflamación , Antiinflamatorios
7.
Biochemistry ; 62(17): 2530-2540, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37540799

RESUMEN

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.


Asunto(s)
Amiloidosis , Glutamina , Humanos , Péptidos , Quimiocinas/química , Membrana Celular/metabolismo , Dicroismo Circular , Receptores CXCR4/metabolismo
8.
J Med Chem ; 66(11): 7070-7085, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37212620

RESUMEN

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.


Asunto(s)
Quimiocinas , Receptores de Quimiocina , Quimiocinas/química , Unión Proteica , Receptores CCR5/metabolismo , Receptores CCR1/metabolismo
9.
Histopathology ; 82(3): 495-503, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36345263

RESUMEN

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.


Asunto(s)
Enfermedad de Hodgkin , Linfadenopatía , Timo , Humanos , Quimiocinas/química , Quimiocinas/inmunología , Enfermedad de Hodgkin/diagnóstico , Enfermedad de Hodgkin/patología , Inmunohistoquímica , Linfadenopatía/patología , Células de Reed-Sternberg/patología , Microambiente Tumoral , Timo/inmunología , Timo/metabolismo
10.
J Struct Biol ; 214(3): 107877, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35750237

RESUMEN

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.


Asunto(s)
Receptores de Quimiocina , Transducción de Señal , Sitios de Unión , Quimiocinas/química , Quimiocinas/metabolismo , Unión Proteica , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo
11.
Chem Commun (Camb) ; 58(26): 4132-4148, 2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35274633

RESUMEN

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.


Asunto(s)
Quimiocinas , Descubrimiento de Drogas , Sitio Alostérico , Sitios de Unión , Quimiocinas/química , Quimiocinas/metabolismo
12.
J Med Chem ; 64(20): 15349-15366, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34662112

RESUMEN

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.


Asunto(s)
Péptidos Antimicrobianos/farmacología , Antituberculosos/farmacología , Quimiocinas/química , Lisina/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Péptidos Antimicrobianos/síntesis química , Péptidos Antimicrobianos/química , Antituberculosos/síntesis química , Antituberculosos/química , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Eritrocitos/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Infecciones por Pseudomonas/tratamiento farmacológico , Estereoisomerismo , Relación Estructura-Actividad , Células THP-1
14.
Cell Mol Life Sci ; 78(17-18): 6265-6281, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34241650

RESUMEN

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.


Asunto(s)
Ligandos , Receptores Acoplados a Proteínas G/metabolismo , Arrestinas/metabolismo , Sitios de Unión , Quimiocinas/química , Quimiocinas/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Inmunidad Innata , Microscopía Confocal , Simulación del Acoplamiento Molecular , Mutagénesis , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Quimiocina/química , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética
15.
Nat Commun ; 12(1): 4151, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34230484

RESUMEN

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.


Asunto(s)
Quimiocinas/química , Quimiocinas/metabolismo , Receptores CCR5/química , Receptores CCR5/metabolismo , Sitios de Unión , Quimiocina CCL3/metabolismo , Quimiocina CCL5/química , Quimiocina CCL5/metabolismo , Microscopía por Crioelectrón , Ligandos , Modelos Moleculares , Conformación Proteica , Receptores CCR5/genética
16.
J Med Chem ; 64(6): 3048-3058, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33705662

RESUMEN

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.


Asunto(s)
Quimiocinas/química , Quimiocinas/farmacología , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Receptores de Quimiocina/agonistas , Animales , Sitios de Unión , Bovinos , Descubrimiento de Drogas , Humanos , Ratones , Simulación del Acoplamiento Molecular , Conformación Proteica , Receptores de Quimiocina/metabolismo
18.
Front Immunol ; 11: 1629, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849553

RESUMEN

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.


Asunto(s)
Amiloide/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Amiloide/química , Proteínas Amiloidogénicas/química , Proteínas Amiloidogénicas/metabolismo , Animales , Antiinfecciosos/química , Antiinfecciosos/farmacología , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Biomarcadores , Quimiocinas/química , Quimiocinas/farmacología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad , Inmunidad Innata , Factores Inmunológicos/química , Factores Inmunológicos/farmacología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Ligandos , Simulación de Dinámica Molecular , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Proteínas Citotóxicas Formadoras de Poros/química , Agregado de Proteínas , Agregación Patológica de Proteínas/metabolismo , Conformación Proteica , Relación Estructura-Actividad , Receptores Toll-Like/química , Receptores Toll-Like/metabolismo
19.
Handb Exp Pharmacol ; 262: 231-258, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32661663

RESUMEN

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.


Asunto(s)
Quimiocinas/química , Infecciones por VIH , Huesos , Quimiocinas/inmunología , Humanos , Osteoblastos/química , Osteoblastos/fisiología , Osteoclastos/química , Osteoclastos/fisiología
20.
Exp Biol Med (Maywood) ; 245(16): 1518-1528, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32715782

RESUMEN

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.


Asunto(s)
Quimiocinas/metabolismo , Enfermedad , Terapia Molecular Dirigida , Animales , Quimiocinas/química , Quimiocinas/genética , Quimiotaxis , Humanos , Modelos Biológicos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA