Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Trends Biochem Sci ; 45(2): 108-122, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31679840

RESUMEN

Ticks are hematophagous arachnids that parasitize mammals and other hosts, feeding on their blood. Ticks secrete numerous salivary factors that enhance host blood flow or suppress the host inflammatory response. The recruitment of leukocytes, a hallmark of inflammation, is regulated by chemokines, which activate chemokine receptors on the leukocytes. Ticks target this process by secreting glycoproteins called Evasins, which bind to chemokines and prevent leukocyte recruitment. This review describes the recent discovery of numerous Evasins produced by ticks, their classification into two structural and functional classes, and the efficacy of Evasins in animal models of inflammatory diseases. The review also proposes a standard nomenclature system for Evasins and discusses the potential of repurposing or engineering Evasins as therapeutic anti-inflammatory agents.


Asunto(s)
Quimiocinas/antagonistas & inhibidores , Proteínas de Insectos/metabolismo , Proteínas y Péptidos Salivales/metabolismo , Garrapatas/metabolismo , Animales , Leucocitos/metabolismo , Receptores de Quimiocina/metabolismo , Terminología como Asunto
2.
J Biol Chem ; 298(10): 102382, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35973511

RESUMEN

Class A tick evasins are natural chemokine-binding proteins that block the signaling of multiple chemokines from the CC subfamily through their cognate receptors, thus suppressing leukocyte recruitment and inflammation. Development of tick evasins as chemokine-targeted anti-inflammatory therapeutics requires an understanding of the factors controlling their chemokine recognition and selectivity. To investigate the role of the evasin N-terminal region for chemokine recognition, we prepared chimeric evasins by interchanging the N-terminal regions of four class A evasins, including a newly identified evasin, EVA-RPU02. We show through chemokine binding analysis of the parental and chimeric evasins that the N-terminal region is critical for chemokine binding affinity and selectivity. Notably, we found some chimeras were unable to bind certain cognate chemokine ligands of both parental evasins. Moreover, unlike any natural evasins characterized to date, some chimeras exhibited specific binding to a single chemokine. These results indicate that the evasin N terminus interacts cooperatively with the "body" of the evasin to enable optimum chemokine recognition. Furthermore, the altered chemokine selectivity of the chimeras validates the approach of engineering the N termini of evasins to yield unique chemokine recognition profiles.


Asunto(s)
Proteínas de Artrópodos , Quimiocinas , Receptores CXCR , Rhipicephalus , Proteínas y Péptidos Salivales , Animales , Proteínas de Artrópodos/metabolismo , Quimiocinas/metabolismo , Unión Proteica , Receptores CXCR/metabolismo , Rhipicephalus/metabolismo , Transducción de Señal , Proteínas y Péptidos Salivales/metabolismo
3.
Immunology ; 165(3): 355-368, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34964126

RESUMEN

Mucositis is a major clinical complication associated with cancer treatment and may limit the benefit of chemotherapy. Leukocytes and inflammatory mediators have been extensively associated with mucositis severity. However, the role of eosinophils in the pathophysiology of chemotherapy-induced mucositis remains to be elucidated. Here, using GATA-1-deficient mice, we investigated the role of eosinophils in intestinal mucositis. There was marked accumulation of eosinophils in mice given irinotecan and eosinophil ablation inhibited intestinal mucositis. Treatment with Evasin-4, a chemokine receptor antagonist, reduced the recruitment of eosinophils and decreased irinotecan-induced mucositis. Importantly, Evasin-4 did not interfere negatively with the antitumour effects of irinotecan. Evasin-4 was of benefit for mice given high doses of irinotecan once Evasin-4-treated mice presented delayed mortality. Altogether, our findings suggest that Evasin-4 may have significant mucosal-protective effects in the context of antineoplastic chemotherapy and may, therefore, be useful in combination with anticancer treatment in cancer patients.


Asunto(s)
Antineoplásicos , Mucositis , Animales , Antineoplásicos/uso terapéutico , Camptotecina/efectos adversos , Eosinófilos/patología , Humanos , Mucosa Intestinal/patología , Irinotecán/efectos adversos , Ratones , Mucositis/inducido químicamente , Mucositis/tratamiento farmacológico , Mucositis/patología
4.
J Biol Chem ; 295(32): 10926-10939, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32471866

RESUMEN

Chemokines mediate leukocyte migration and homeostasis and are key targets in inflammatory diseases including atherosclerosis, cytokine storm, and chronic autoimmune disease. Chemokine redundancy and ensuing network robustness has frustrated therapeutic development. Salivary evasins from ticks bind multiple chemokines to overcome redundancy and are effective in several preclinical disease models. Their clinical development has not progressed because of concerns regarding potential immunogenicity, parenteral delivery, and cost. Peptides mimicking protein activity can overcome the perceived limitations of therapeutic proteins. Here we show that peptides possessing multiple chemokine-binding and anti-inflammatory activities can be developed from the chemokine-binding site of an evasin. We used hydrogen-deuterium exchange MS to map the binding interface of the evasin P672 that physically interacts with C-C motif chemokine ligand (CCL) 8 and synthesized a 16-mer peptide (BK1.1) based on this interface region in evasin P672. Fluorescent polarization and native MS approaches showed that BK1.1 binds CCL8, CCL7, and CCL18 and disrupts CCL8 homodimerization. We show that a BK1.1 derivative, BK1.3, has substantially improved ability to disrupt P672 binding to CCL8, CCL2, and CCL3 in an AlphaScreen assay. Using isothermal titration calorimetry, we show that BK1.3 directly binds CCL8. BK1.3 also has substantially improved ability to inhibit CCL8, CCL7, CCL2, and CCL3 chemotactic function in vitro We show that local as well as systemic administration of BK1.3 potently blocks inflammation in vivo Identification and characterization of the chemokine-binding interface of evasins could thus inspire the development of novel anti-inflammatory peptides that therapeutically target the chemokine network in inflammatory diseases.


Asunto(s)
Antiinflamatorios/química , Quimiocina CCL8/metabolismo , Péptidos/química , Ingeniería de Proteínas , Receptores de Quimiocina/metabolismo , Secuencia de Aminoácidos , Animales , Antiinflamatorios/farmacología , Dimerización , Humanos , Espectrometría de Masas/métodos , Péptidos/farmacología , Unión Proteica , Homología de Secuencia de Aminoácido , Garrapatas/metabolismo
5.
J Pept Sci ; 27(2): e3290, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33118239

RESUMEN

Thiazolidine ring-opening reaction is one of the key steps in protein chemical synthesis via sequential native chemical ligation strategy. We recently developed a novel thiazolidine ring-opening reaction with 2,2'-dipyridyl disulfide (DPDS). In order to investigate the applicability of this reaction to glycoprotein synthesis, we synthesized evasin-3, a cysteine-rich glycoprotein with chemokine-binding ability originally found in tick saliva. The sequence of evasin-3 was divided into three segments, and these segments were separately synthesized with the ordinary solid-phase peptide synthesis method. After the first ligation of middle and C-terminal segments, thiazolidine used as a protecting group of Cys residue at the N-terminus of the middle segment was converted to Cys with DPDS. In this thiazolidine ring-opening reaction, DPDS treatment did not affect the N-linked glycan moiety. After the second ligation with the N-terminal segment and the refolding reaction, evasin-3 could be obtained in good yield. The synthetic evasin-3 showed the binding ability specifically to CXCL chemokines. These results clearly indicate that this DPDS method is useful for glycoprotein synthesis.


Asunto(s)
2,2'-Dipiridil/análogos & derivados , Proteínas de Artrópodos/síntesis química , Disulfuros/química , Proteínas y Péptidos Salivales/síntesis química , Tiazolidinas/química , 2,2'-Dipiridil/química , Proteínas de Artrópodos/química , Estructura Molecular , Receptores CXCR/química , Proteínas y Péptidos Salivales/química
6.
J Biol Chem ; 294(29): 11199-11212, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31167786

RESUMEN

Tick evasins (EVAs) bind either CC- or CXC-chemokines by a poorly understood promiscuous or "one-to-many" mechanism to neutralize inflammation. Because EVAs potently inhibit inflammation in many preclinical models, highlighting their potential as biological therapeutics for inflammatory diseases, we sought to further unravel the CXC-chemokine-EVA interactions. Using yeast surface display, we identified and characterized 27 novel CXC-chemokine-binding evasins homologous to EVA3 and defined two functional classes. The first, which included EVA3, exclusively bound ELR+ CXC-chemokines, whereas the second class bound both ELR+ and ELR- CXC-chemokines, in several cases including CXC-motif chemokine ligand 10 (CXCL10) but, surprisingly, not CXCL8. The X-ray crystal structure of EVA3 at a resolution of 1.79 Å revealed a single antiparallel ß-sheet with six conserved cysteine residues forming a disulfide-bonded knottin scaffold that creates a contiguous solvent-accessible surface. Swapping analyses identified distinct knottin scaffold segments necessary for different CXC-chemokine-binding activities, implying that differential ligand positioning, at least in part, plays a role in promiscuous binding. Swapping segments also transferred chemokine-binding activity, resulting in a hybrid EVA with dual CXCL10- and CXCL8-binding activities. The solvent-accessible surfaces of the knottin scaffold segments have distinctive shape and charge, which we suggest drives chemokine-binding specificity. These studies provide structural and mechanistic insight into how CXC-chemokine-binding tick EVAs achieve class specificity but also engage in promiscuous binding.


Asunto(s)
Quimiocinas CXC/metabolismo , Miniproteínas Nodales de Cistina/metabolismo , Receptores de Quimiocina/metabolismo , Garrapatas/metabolismo , Animales , Cristalografía por Rayos X , Unión Proteica , Conformación Proteica , Receptores de Quimiocina/genética , Receptores de Quimiocina/aislamiento & purificación , Especificidad de la Especie , Garrapatas/clasificación , Levaduras/genética
7.
J Biol Chem ; 293(16): 6134-6146, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29487134

RESUMEN

Tick chemokine-binding proteins (evasins) are an emerging class of biologicals that target multiple chemokines and show anti-inflammatory activities in preclinical disease models. Using yeast surface display, we identified a CCL8-binding evasin, P672, from the tick Rhipicephalus pulchellus We found that P672 binds CCL8 and eight other CC-class chemokines with a Kd < 10 nm and four other CC chemokines with a Kd between 10 and 100 nm and neutralizes CCL3, CCL3L1, and CCL8 with an IC50 < 10 nm The CC chemokine-binding profile was distinct from that of evasin 1 (EVA1), which does not bind CCL8. We also show that P672's binding activity can be markedly modulated by the location of a StrepII-His purification tag. Combining native MS and bottom-up proteomics, we further demonstrated that P672 is glycosylated and forms a 1:1 complex with CCL8, disrupting CCL8 homodimerization. Homology modeling of P672 using the crystal structure of the EVA1 and CCL3 complex as template suggested that 44 N-terminal residues of P672 form most of the contacts with CCL8. Replacing the 29 N-terminal residues of EVA1 with the 44 N-terminal residues of P672 enabled this hybrid evasin to bind and neutralize CCL8, indicating that the CCL8-binding properties of P672 reside, in part, in its N-terminal residues. This study shows that the function of certain tick evasins can be manipulated simply by adding a tag. We conclude that homology modeling helps identify regions with transportable chemokine-binding functions within evasins, which can be used to construct hybrid evasins with altered properties.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Quimiocinas/metabolismo , Receptores de Quimiocina/metabolismo , Garrapatas/metabolismo , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Glicosilación , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Receptores de Quimiocina/química , Receptores de Quimiocina/genética , Saccharomyces cerevisiae/genética , Espectrometría de Masas en Tándem
8.
J Biol Chem ; 292(38): 15670-15680, 2017 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-28778927

RESUMEN

To prolong residence on their hosts, ticks secrete many salivary factors that target host defense molecules. In particular, the tick Rhipicephalus sanguineus has been shown to produce three salivary glycoproteins named "evasins," which bind to host chemokines, thereby inhibiting the recruitment of leukocytes to the location of the tick bite. Using sequence similarity searches, we have identified 257 new putative evasin sequences encoded by the genomes or salivary or visceral transcriptomes of numerous hard ticks, spanning the genera Rhipicephalus, Amblyomma, and Ixodes of the Ixodidae family. Nine representative sequences were successfully expressed in Escherichia coli, and eight of the nine candidates exhibited high-affinity binding to human chemokines. Sequence alignments enabled classification of the evasins into two subfamilies: C8 evasins share a conserved set of eight Cys residues (four disulfide bonds), whereas C6 evasins have only three of these disulfide bonds. Most of the identified sequences contain predicted secretion leader sequences, N-linked glycosylation sites, and a putative site of tyrosine sulfation. We conclude that chemokine-binding evasin proteins are widely expressed among tick species of the Ixodidae family, are likely to play important roles in subverting host defenses, and constitute a valuable pool of anti-inflammatory proteins for potential future therapeutic applications.


Asunto(s)
Quimiocinas/antagonistas & inhibidores , Ixodidae/genética , Receptores de Quimiocina/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Bases de Datos Genéticas , Escherichia coli/genética , Evolución Molecular , Genómica , Ixodidae/clasificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Quimiocina/química , Receptores de Quimiocina/genética , Alineación de Secuencia
9.
Front Cell Infect Microbiol ; 11: 769542, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34746035

RESUMEN

Chemokines are structurally related proteins that activate leucocyte migration in response to injury or infection. Tick saliva contains chemokine-binding proteins or evasins which likely neutralize host chemokine function and inflammation. Biochemical characterisation of 50 evasins from Ixodes, Amblyomma and Rhipicephalus shows that they fall into two functional classes, A and B, with exclusive binding to either CC- or CXC- chemokines, respectively. Class A evasins, EVA1 and EVA4 have a four-disulfide-bonded core, whereas the class B evasin EVA3 has a three-disulfide-bonded "knottin" structure. All 29 class B evasins have six cysteine residues conserved with EVA3, arrangement of which defines a Cys6-motif. Nineteen of 21 class A evasins have eight cysteine residues conserved with EVA1/EVA4, the arrangement of which defines a Cys8-motif. Two class A evasins from Ixodes (IRI01, IHO01) have less than eight cysteines. Many evasin-like proteins have been identified in tick salivary transcriptomes, but their phylogenetic relationship with respect to biochemically characterized evasins is not clear. Here, using BLAST searches of tick transcriptomes with biochemically characterized evasins, we identify 292 class A and 157 class B evasins and evasin-like proteins from Prostriate (Ixodes), and Metastriate (Amblyomma, Dermacentor, Hyalomma, Rhipicephalus) ticks. Phylogenetic analysis shows that class A evasins/evasin-like proteins segregate into two classes, A1 and A2. Class A1 members are exclusive to Metastriate ticks and typically have a Cys8-motif and include EVA1 and EVA4. Class A2 members are exclusive to Prostriate ticks, lack the Cys8-motif, and include IHO01 and IRI01. Class B evasins/evasin-like proteins are present in both Prostriate and Metastriate lineages, typically have a Cys6-motif, and include EVA3. Most evasins/evasin-like proteins in Metastriate ticks belong to class A1, whereas in Prostriate species they are predominantly class B. In keeping with this, the majority of biochemically characterized Metastriate evasins bind CC-chemokines, whereas the majority of Prostriate evasins bind CXC-chemokines. While the origin of the structurally dissimilar classes A1 and A2 is yet unresolved, these results suggest that class B evasin-like proteins arose before the divergence of Prostriate and Metastriate lineages and likely functioned to neutralize CXC-chemokines and support blood feeding.


Asunto(s)
Proteínas de Artrópodos/clasificación , Ixodidae , Receptores de Quimiocina/clasificación , Proteínas y Péptidos Salivales/clasificación , Garrapatas , Animales , Quimiocinas/metabolismo , Ixodidae/genética , Ixodidae/metabolismo , Filogenia , Unión Proteica , Garrapatas/metabolismo
10.
mBio ; 12(6): e0305921, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34872352

RESUMEN

Rickettsiae are obligate intracellular Gram-negative bacteria transmitted by arthropod vectors. Despite their reduced genomes, the function(s) of the majority of rickettsial proteins remains to be uncovered. APRc is a highly conserved retropepsin-type protease, suggested to act as a modulator of other rickettsial surface proteins with a role in adhesion/invasion. However, APRc's function(s) in bacterial pathogenesis and virulence remains unknown. This study demonstrates that APRc targets host serum components, combining nonimmune immunoglobulin (Ig)-binding activity with resistance to complement-mediated killing. We confirmed nonimmune human IgG binding in extracts of different rickettsial species and intact bacteria. Our results revealed that the soluble domain of APRc is capable of binding to human (h), mouse, and rabbit IgG and different classes of human Ig (IgG, IgM, and IgA) in a concentration-dependent manner. APRc-hIgG interaction was confirmed with total hIgG and normal human serum. APRc-hIgG displayed a binding affinity in the micromolar range. We provided evidence of interaction preferentially through the Fab region and confirmed that binding is independent of catalytic activity. Mapping the APRc region responsible for binding revealed the segment between amino acids 157 and 166 as one of the interacting regions. Furthermore, we demonstrated that expression of the full-length protease in Escherichia coli is sufficient to promote resistance to complement-mediated killing and that interaction with IgG contributes to serum resistance. Our findings position APRc as a novel Ig-binding protein and a novel moonlighting immune evasion factor of Rickettsia, contributing to the arsenal of virulence factors utilized by these intracellular pathogens to aid in host colonization. IMPORTANCE Many Rickettsia organisms are pathogenic to humans, causing severe infections, like Rocky Mountain spotted fever and Mediterranean spotted fever. However, immune evasion mechanisms and pathogenicity determinants in rickettsiae are far from being resolved. We provide evidence that the highly conserved rickettsial retropepsin-type protease APRc displays nonimmune immunoglobulin (Ig)-binding activity and participates in serum resistance. APRc emerges then as a novel Ig-binding protein from Gram-negative bacteria and the first to be identified in Rickettsia. Bacterial surface proteins capable of Ig binding are known to be multifunctional and key players in immune evasion. We demonstrate that APRc is also a novel moonlighting protein, exhibiting different actions on serum components and acting as a novel evasin. This work strengthens APRc as a virulence factor in Rickettsia and its significance as a potential therapeutic target. Our findings significantly contribute to a deeper understanding of the virulence strategies used by intracellular pathogens to subvert host immune responses.


Asunto(s)
Proteínas Bacterianas/inmunología , Evasión Inmune , Inmunoglobulinas/inmunología , Péptido Hidrolasas/inmunología , Rickettsia/inmunología , Fiebre Maculosa de las Montañas Rocosas/inmunología , Animales , Proteínas Bacterianas/genética , Proteínas del Sistema Complemento/inmunología , Humanos , Ratones , Péptido Hidrolasas/química , Péptido Hidrolasas/genética , Dominios Proteicos , Conejos , Rickettsia/genética , Fiebre Maculosa de las Montañas Rocosas/microbiología
11.
Artículo en Inglés | MEDLINE | ID: mdl-31997766

RESUMEN

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.


Asunto(s)
Quimiocinas/metabolismo , Inflamación/metabolismo , Proteínas de Insectos/metabolismo , Animales , Quimiocinas/química , Humanos , Peptidomiméticos
12.
Methods Enzymol ; 570: 187-205, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26921947

RESUMEN

Chemokines are small chemoattractant proteins involved in the recruitment of leukocytes to the site of inflammation. Due to their prominent role in the inflammatory process, chemokine inhibitors have been developed by parasites to remain undetected not only by the host immune system but also by various laboratories to develop anti-inflammatory compounds. Taking advantage of the small size of natural chemokine-binding proteins, we report here several methods to facilitate their characterization using phage display to identify the chemokine-binding site and to modulate the selectivity of such inhibitors. Interestingly, these methods could be adapted to display the natural inhibitors of other cytokines or even cytokines on phage surface.


Asunto(s)
Técnicas de Visualización de Superficie Celular/métodos , Quimiocinas/metabolismo , Biología Molecular/métodos , Mutagénesis , Alanina/genética , Sitios de Unión , Proteínas Portadoras/metabolismo , Clonación Molecular , Simulación por Computador , Ensayo de Inmunoadsorción Enzimática , Ligandos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
13.
Front Immunol ; 4: 193, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874339

RESUMEN

The CC chemokine ligand 18 (CCL18) was first identified as a chemoattractant for naïve T cells. It has been reported to recruit T and B lymphocytes, and we show here, natural killer (NK) cells, but with low efficacy. Investigation of its ability to elicit G-protein-coupled signaling showed that it does not involve extracellular signal-regulated kinase (ERK) phosphorylation, and it is not able to induce receptor internalization, as assessed on CCR3. CCL18 has recently been reported to possess activities unrelated to cellular recruitment, but it had no effect on T lymphocyte proliferation. We postulated that a more potent chemoattractant may be produced under inflammatory conditions but only minor truncations were observed, with the major form being the full-length protein. In view of the lack of potent immunomodulatory properties, we wondered if binding to CCL18 by the tick chemokine binding proteins Evasin-1 and -4 was an artifact of the methods used, but complex formation was confirmed by size exclusion chromatography, and abrogation of its binding to, and antagonism of, CCR3. Its receptor has remained elusive since its cloning in 1997, although it has been reported to induce migration of breast cancer cells by signaling through PITPNM3, but we show that this receptor is not expressed on lymphocytes. We have developed a radiolabeled equilibrium competition binding assay and demonstrated that it bound with high affinity to peripheral blood leukocytes (PBLs), but the binding was displaced similarly by both unlabelled CCL18 as well as heparin. Both heparin binding and binding to PBLs are considerably abrogated by mutation of the BBXB motif in the 40s loop suggesting an essential role of the CCL18-glycosaminoglycan interaction.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA