Synthetic Immunology

We are applying a number of genome engineering tools (e.g., CRISPR-Cas9) to reprogram immune cells for applications related to biotechnology and cellular immunotherapy.

Synthetic_Figure_0

Reprogramming immune cells for protein expression and engineering

In one of our research areas, we focus on the development of technologies for protein expression and engineering using advanced methods in immunogenomic reprogramming.

Specifically, we have recently developed Plug-and-(dis)play hybridoma cells, which is a platform for rapid reprogramming of antibody specificity by immunogenomic engineering (Pogson*, Parola* et al., Nature Communications, 2016).

Hybrdomas are stable, rapidly-proliferating cell lines widely utilized for antibody screening and production, which are generated by fusing primary mouse B cells with myelomas tumor cells. Antibody specificity of a hybridoma clone is determined by the immunoglobulin sequence of the primary B cell. We use CRISPR-Cas9 to generate double-stranded breaks in immunoglobulin loci, enabling deletion of the native variable light chain and replacement of the endogenous variable heavy chain with a fluorescent reporter protein (mRuby) (Fig. 1). New antibody genes are introduced by Cas9-targeting of mRuby for replacement with a donor construct encoding a light chain and a variable heavy chain, resulting in full-length antibody expression. Since hybridomas surface express and secrete antibodies, reprogrammed cells are isolated using flow cytometry and cell culture supernatant is used for antibody production. Plug-and-(dis)play hybridomas can be reprogrammed with only a single transfection and screening step. 

Synthetic_Figure_1
Figure 1. Reprogramming of hybridomas’ antibody specificity via Plug-and-(dis)play. (a) Generation of the Plug-and-(dis)play(PnP)-mRuby cell line. The antibody variable heavy (VH) region of the parental hybridoma cell line is targeted for CRISPR-Cas9 mediated homology directed repair (HDR) to replace the endogenous VH with the fluorescent reporter mRuby; the endogenous variable light (VL) region is deleted via multiplexed targeting by CRISPR-Cas9 and consequent non-homologous end joining (NHEJ). (b) Generation of cell lines stably expressing full-length synthetic antibodies. Analogously to the cassette exchange to replace VH with mRuby, the fluorescent reporter is now replaced by a HDR donor containing: variable light chain, constant light region (CK), 2A peptide for ribosomal skipping and synthesis of two separate proteins from a single open reading frame, and variable heavy chain. Recombination donors are provided with the same homology arms as the Ruby template. Due to targeted integration in the VH locus, splicing with the heavy constant region (CH) will result in a single open reading frame encoding for a full-length antibody. Since the constant region is necessary for the antibody to be surface expressed, FACS staining functions as a selection marker for targeted integration. 

Reprogramming immune cells for cellular immunotherapy

The rapid advances in genome editing technologies are stimulating tremendous excitement for developing advanced cellular immunotherapies. To date, proof-of-concept examples of genome editing in immune cells have shown targeted insertion, deletion, or point mutation of genes. However, what has yet to be reported is the exchange of entire genes, particularly those of polymorphic immunogenomic loci. The ability to exchange immunogenomic alleles would enable the reprogramming of immune cell specificity.

One important immunogenomic region is the MHC locus; these genes are critically important in allogeneic hematopoietic stem cell (HSC) transplantation procedures, where MHC matching between the donor and the host is a primary determinant of transplant success. While the editing of human HSCs has traditionally been difficult, new advances are greatly improving efficacy to the levels required for clinical application. In fact, very recent developments indicate genome editing of human HSCs will shortly gain clinical approval in Europe for Severe Combined Immune Deficiency disease. However, what remains a major challenge is the need to still obtain proper MHC matching of donor and host.

As a proof of concept for MHC reprogramming, we targeted a murine MHC locus (H2-K1) with our approach of CRISPR-Cas9 assisted cassette exchange (Kelton et al., Scientific Reports, 2017).

In murine-derived antigen-presenting cell lines, we introduced double strand breaks with a multiplex-targeting Cas9 approach. Providing repair templates, we enabled homology directed repair (HDR) of the double-stranded break. Thus, we were able to effectively replace a MHC-I allele (H2-Kd) of ~3.4 kb with a highly similar orthogonal allele (H2-Kb). We validated the altered cellular phenotype by the surface expression of the new MHC alleles, using fluorescence associated cell sorting(and a T cell activation assay, highlighting functional immune activity of the MHC exchanged cells.

Synthetic_Figure_2
Figure 2. Schematic outline of our experimental approach for the exchange of MHC alleles by CACE. Immortalized murine cells possessing the H2-Kd genotype are cleaved by Cas9 in the presence of a donor template generated from a murine cell line carrying a H2-Kb genotype. HDR mediates seamless exchange of the template allowing the presentation of a certain, H2-Kb restricted peptide.
JavaScript has been disabled in your browser