Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Exp Allergy ; 53(6): 636-647, 2023 06.
Article in English | MEDLINE | ID: mdl-37038893

ABSTRACT

BACKGROUND: Clinical and experimental analyses indicate a pathognomonic role for allergen IgE crosslinking through epitope-paratope interactions as a major initial step in the cascade leading to effector cell activation and clinical manifestations of lgE-mediated food allergies. We aimed to undertake the initial development and assessment of Ara h 2-specific IgE epitope-like peptides that can bind to allergen-specific IgE paratopes and suppress effector cell activation. METHODS: We performed biopanning, screening, IgE binding, selection and mapping of peptides. We generated synthetic peptides for use in all functional experiments. ImmunoCAP inhibition, basophil and mast cell activation tests, with LAD2 cells, a human mast cell line were performed. Twenty-six children or young adults who had peanut allergy were studied. RESULTS: We identified and selected three linear peptides (DHPRFNRDNDVA, DHPRYGP and DHPRFST), and immunoblot analyses revealed binding to lgE from peanut-allergic individuals. The peptide sequences were aligned to the disordered region corresponding to the loop between helices 2 and 3 of Ara h 2, and conformational mapping showed that the peptides match the surface of Ara h 2 and h 6 but not other peanut allergens. In ImmunoCAP inhibition experiments, the peptides significantly inhibit the binding of IgE to Ara h 2 (p < .001). In basophil and mast cell activation tests, the peptides significantly suppressed Ara h 2-induced effector cell activation (p < .05) and increased the half-maximal Ara h 2 effective concentration (p < .05). Binding of the peptides to specific IgEs did not induce activation of basophils or mast cells. CONCLUSIONS: These studies show that the indicated peptides reduce the allergenic activity of Ara h 2 and suppress lgE-dependent basophil and mast cell activation. These observations may suggest a novel therapeutic strategy for food allergy based on epitope-paratop blocking.


Subject(s)
Food Hypersensitivity , Peanut Hypersensitivity , Child , Young Adult , Humans , Epitopes , Antigens, Plant , Glycoproteins , Peptides , Immunoglobulin E , Allergens , Arachis , 2S Albumins, Plant
2.
Clin Rev Allergy Immunol ; 58(3): 326-341, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31240545

ABSTRACT

Bee venom immunotherapy is the main treatment option for bee sting allergy. Its major limitations are the high percentage of allergic side effects and long duration, which are driving the development of novel therapeutic modalities. Three general approaches have been evaluated including the use of hypoallergenic allergen derivatives, adjunctive therapy, and alternative delivery routes. This article reviews preclinical and clinical evidence on the therapeutic potential of these new therapies. Among hypoallergenic derivatives, hybrid allergens showed a markedly reduced IgE reactivity in mouse models. Whether they will offer therapeutic benefit over extract, it is still not known since clinical trials have not been carried out yet. T cell epitope peptides have proven effective in small clinical trials. Major histocompatibility complex class II restriction was circumvented by using long overlapping or promiscuous T cell epitope peptides. However, the T cell-mediated late-phase adverse events have been reported with both short and longer peptides. Application of mimotopes could potentially overcome both T cell- and IgE-mediated adverse events. During this evolution of vaccine, there has been a gain in safety. The efficacy was further improved with the use of Toll-like receptor-activating adjuvants and delivery systems. In murine models, the association of allergen Api m 1 with cytosine-guanosine rich oligonucleotides stimulated strong T-helper type-1 response, whereas its encapsulation into microbubbles protected mice against allergen challenge. An intralymphatic administration of low-dose vaccine has shown the potential to decrease treatment from 5 years to only 12 weeks. Bigger clinical trials are needed to follow up on these results.


Subject(s)
Allergens/immunology , Bee Venoms/immunology , Desensitization, Immunologic/methods , Epitopes/immunology , Hypersensitivity/therapy , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Animals , Bees , Desensitization, Immunologic/trends , Humans , Hypersensitivity/immunology , Hypersensitivity, Immediate , Mice
3.
Mol Immunol ; 71: 176-183, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26908079

ABSTRACT

Cat allergy is one of the most prevalent allergies worldwide and can lead to the development of rhinitis and asthma. Thus far, only allergen extracts from natural sources have been used for allergen-specific immunotherapy. However, extracts and whole allergens in immunotherapy present an anaphylaxis risk. Identification of allergen epitopes or mimotopes has an important role in development of safe and effective allergen-specific immunotherapy. Moreover, with a suitable immunogenic carrier, the absence of sufficient immune response elicited by short peptides could be surmounted. In this study, we identified five structural mimotopes of the major cat allergen Fel d 1 by immunoscreening with random peptide phage libraries. The mimotopes were computationally mapped to the allergen surface, and their IgE reactivity was confirmed using sera from cat-allergic patients. Importantly, the mimotopes showed no basophil activation of the corresponding cat-allergic patients, which makes them good candidates for the development of hypoallergenic vaccine. As bacteriophage particles are becoming increasingly recognized as immunogenic carriers, we constructed bacteriophage particles displaying multiple copies of each selected mimotope on major phage coat protein. These constructed phages elicited T cell-mediated immune response, which was predominated by the type 1 T cell response. Mimotopes alone contributed to the type 1 T cell response by promoting IL-2 production. Fel d 1 mimotopes, as well as their filamentous phage immunogenic carriers, represent promising candidates in the development of hypoallergenic vaccine against cat allergy.


Subject(s)
Desensitization, Immunologic/methods , Glycoproteins/immunology , Hypersensitivity/prevention & control , Adult , Allergens/immunology , Animals , Basophil Degranulation Test , Blotting, Western , Cats , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Genetic Vectors , Humans , Immunoassay , Inovirus , Molecular Mimicry , Peptide Library
4.
Eur J Med Chem ; 75: 308-26, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24556146

ABSTRACT

Dendritic cell-specific, intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) is a C-type lectin expressed specifically on dendritic cells. It is a primary site for recognition and binding of various pathogens and thus a promising therapeutic target for inhibition of pathogen entry and subsequent prevention of immune defense cell infection. We report the design and synthesis of d-mannose-based DC-SIGN antagonists bearing diaryl substituted 1,3-diaminopropanol or glycerol moieties incorporated to target the hydrophobic groove of the receptor. The designed glycomimetics were evaluated by in vitro assay of the isolated DC-SIGN extracellular domain for their ability to compete with HIV-1 gp120 for binding to the DC-SIGN carbohydrate recognition domain. Compounds 14d and 14e, that display IC50 values of 40 µM and 50 µM, are among the most potent monovalent DC-SIGN antagonists reported. The antagonistic effect of all the synthesized compounds was further evaluated by a one-point in vitro assay that measures DC adhesion. Compounds 14d, 14e, 18d and 18e were shown to act as functional antagonists of DC-SIGN-mediated DC adhesion. The binding mode of 14d was also studied by molecular docking and molecular dynamics simulation, which revealed flexibility of 14d in the binding site and provides a basis for further optimization.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , HIV Envelope Protein gp120/metabolism , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/metabolism , Mannose/analogs & derivatives , Mannose/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Binding Sites , Cell Adhesion Molecules/chemistry , Dendritic Cells/drug effects , Dendritic Cells/metabolism , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/virology , Humans , Lectins, C-Type/chemistry , Molecular Docking Simulation , Receptors, Cell Surface/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...