Abstract

Autoimmune diseases such as rheumatoid arthritis are caused by immune system recognition of self-proteins and subsequent production of effector T cells that recognize and attack healthy tissue. Therapies for these diseases typically utilize broad immune suppression, which can be effective, but which also come with an elevated risk of susceptibility to infection and cancer. T cell recognition of antigens is driven by binding of T cell receptors to peptides displayed on major histocompatibility complex proteins (MHCs) on the cell surface of antigen-presenting cells. Technology for recombinant production of the extracellular domains of MHC proteins and loading with peptides to produce pMHCs has provided reagents for detection of T cell populations, and with the potential for therapeutic intervention. However, production of pMHCs in large quantities remains a challenge and a translational path needs to be established. Here, we demonstrate a fusion protein strategy enabling large-scale production of pMHCs. A peptide corresponding to amino acids 259–273 of collagen II was fused to the N-terminus of the MHC_II beta chain, and the alpha and beta chains were each fused to human IgG4 Fc domains and co-expressed. A tag was incorporated to enable site-specific conjugation. The cytotoxic drug payload, MMAF, was conjugated to the pMHC and potent, peptide-specific killing of T cells that recognize the collagen pMHC was demonstrated with tetramerized pMHC-MMAF conjugates. Finally, these pMHCs were incorporated into MMAF-loaded 3DNA nanomaterials in order to provide a biocompatible platform. Loading and pMHC density were optimized, and peptide-specific T cell killing was demonstrated. These experiments highlight the potential of a pMHC fusion protein-targeted, drug-loaded nanomaterial approach for selective delivery of therapeutics to disease-relevant T cells and new treatment options for autoimmune disease.

Highlights

  • Another approach that has been demonstrated is coating of the Peptide-loaded MHC complexes (pMHCs) onto nanoparticles which can potentially be tuned to induce outcomes ranging from T cell depletion to Treg expansion, among others [14,15]

  • Monomers for peptide–major histocompatibility complex proteins (MHCs) class II tetramers have been produced in a variety of systems including insect cells and E. coli

  • The biotinylated pMHCs werenotetramerized with streptavidin-phycoerythrin in formation of either of the chains and mutations need be incorporated into the Fc domain equimolar ratios and binding of the tetramers to cell lines specific for the Col_II peptide (DR4.CII.23.5 and CII.26.B6.18) or the HA peptide (HA.5D3.9) was assessed by flow cytometry

Read more

Summary

A Strategy for Selective Deletion of Autoimmunity-Related T

Goldberg 1, *, Nathan Felix 1 , Michael McCauley 1 , Ryan Eberwine 1 , Lou Casta 2 , Kathleen Haskell 1 , Tricia Lin 1 , Elizabeth Palovick 2,† , Donna Klein 1 , Lori Getts 2,† , Robert Getts 2,† , Mimi Zhou 3 , Pratima Bansal-Pakala 1 and Vadim Dudkin 1,‡. Current address: Code Biotherapeutics, Hatfield, PA 19440, USA. Current address: LARONDE Inc., Cambridge, MA 02142, USA

Introduction
Materials
Construction of Plasmids
Expression and Purification of Fusion Proteins
MMAF Conjugation
Protein Characterization Methods
Generation of T cell Hybridomas
Tetramer Formation and FACS
2.10. Cytotoxicity Assays
Design of pMHC Fc Fusion Proteins
Optimization of Conjugation Scheme
Production of 3DNA-pMHC
Activity
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call