Multivalent Soluble Antigen Arrays Exhibit High Avidity Binding and Modulation of B Cell Receptor-Mediated Signaling to Drive Efficacy against Experimental Autoimmune Encephalomyelitis

Autor: Martin A. Leon, Cory Berkland, Chad J. Pickens, Brittany L. Hartwell
Rok vydání: 2017
Předmět:
0301 basic medicine
Azides
Cell signaling
Encephalomyelitis
Autoimmune
Experimental

Immunoconjugates
Polymers and Plastics
Injections
Subcutaneous

Encephalomyelitis
B-cell receptor
Protein Array Analysis
Receptors
Antigen
B-Cell

chemical and pharmacologic phenomena
Bioengineering
Autoantigens
Article
Cell Line
Biomaterials
Mice
03 medical and health sciences
Antigen
immune system diseases
Cell Adhesion
Immune Tolerance
Materials Chemistry
medicine
Animals
Humans
Calcium Signaling
Hyaluronic Acid
Receptor
Cell adhesion
Autoimmune disease
B-Lymphocytes
Cycloaddition Reaction
Chemistry
Experimental autoimmune encephalomyelitis
medicine.disease
nervous system diseases
Cell biology
030104 developmental biology
Gene Expression Regulation
Biochemistry
Alkynes
lipids (amino acids
peptides
and proteins)

Peptides
Zdroj: Biomacromolecules
ISSN: 1526-4602
1525-7797
Popis: A pressing need exists for antigen-specific immunotherapies (ASIT) that induce selective tolerance in autoimmune disease while avoiding deleterious global immunosuppression. Multivalent soluble antigen arrays (SAgA(PLP:LABL)), consisting of a hyaluronic acid (HA) linear polymer backbone co-grafted with multiple copies of autoantigen (PLP) and cell adhesion inhibitor (LABL) peptides, are designed to induce tolerance to a specific multiple sclerosis (MS) autoantigen. Previous studies established that hydrolyzable SAgA(PLP:LABL), employing a degradable linker to codeliver PLP and LABL, was therapeutic in experimental autoimmune encephalomyelitis (EAE) in vivo and exhibited antigen-specific binding with B cells, targeted the B cell receptor (BCR), and dampened BCR-mediated signaling in vitro. Our results pointed to sustained BCR engagement as the SAgA(PLP:LABL) therapeutic mechanism, so we developed a new version of the SAgA molecule using non-hydrolyzable conjugation chemistry, hypothesizing it would enhance and maintain the molecule’s action at the cell surface to improve efficacy. ‘Click SAgA’ (cSAgA(PLP:LABL)) uses hydrolytically stable covalent conjugation chemistry (Copper-catalyzed Azide-Alkyne Cycloaddition (CuAAC)) rather than a hydrolyzable oxime bond to attach PLP and LABL to HA. We explored cSAgA(PLP:LABL) B cell engagement and modulation of BCR-mediated signaling in vitro through flow cytometry binding and calcium flux signaling assays. Indeed, cSAgA(PLP:LABL) exhibited higher avidity B cell binding and greater dampening of BCR-mediated signaling than hydrolyzable SAgA(PLP:LABL). Furthermore, c SAgA(PLP:LABL) exhibited significantly enhanced in vivo efficacy compared to hydrolyzable SAgA(PLP:LABL), achieving equivalent efficacy at one quarter of the dose. These results indicate that non-hydrolyzable conjugation increased the avidity of cSAgA(PLP:LABL) to drive in vivo efficacy through modulated BCR-mediated signaling.
Databáze: OpenAIRE