1. Academic Validation
  2. Targeted gene editing restores regulated CD40L function in X-linked hyper-IgM syndrome

Targeted gene editing restores regulated CD40L function in X-linked hyper-IgM syndrome

  • Blood. 2016 May 26;127(21):2513-22. doi: 10.1182/blood-2015-11-683235.
Nicholas Hubbard 1 David Hagin 1 Karen Sommer 1 Yumei Song 1 Iram Khan 1 Courtnee Clough 1 Hans D Ochs 2 David J Rawlings 3 Andrew M Scharenberg 3 Troy R Torgerson 2
Affiliations

Affiliations

  • 1 Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA; and.
  • 2 Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA; and Department of Pediatrics and.
  • 3 Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA; and Department of Pediatrics and Department of Immunology, University of Washington, Seattle, WA.
Abstract

Loss of CD40 Ligand (CD40L) expression or function results in X-linked hyper-immunoglobulin (Ig)M syndrome (X-HIGM), characterized by recurrent infections due to impaired immunoglobulin class-switching and somatic hypermutation. Previous attempts using retroviral gene transfer to correct murine CD40L expression restored immune function; however, treated mice developed lymphoproliferative disease, likely due to viral-promoter-dependent constitutive CD40L expression. These observations highlight the importance of preserving endogenous gene regulation in order to safely correct this disorder. Here, we report efficient, on-target, homology-directed repair (HDR) editing of the CD40LG locus in primary human T cells using a combination of a transcription activator-like effector nuclease-induced double-strand break and a donor template delivered by recombinant adeno-associated virus. HDR-mediated insertion of a coding sequence (green fluorescent protein or CD40L) upstream of the translation start site within exon 1 allowed transgene expression to be regulated by endogenous CD40LG promoter/enhancer elements. Additionally, inclusion of the CD40LG 3'-untranslated region in the transgene preserved posttranscriptional regulation. Expression kinetics of the transgene paralleled that of endogenous CD40L in unedited T cells, both at rest and in response to T-cell stimulation. The use of this method to edit X-HIGM patient T cells restored normal expression of CD40L and CD40-murine IgG Fc fusion protein (CD40-muIg) binding, and rescued IgG class switching of naive B cells in vitro. These results demonstrate the feasibility of engineered nuclease-directed gene repair to restore endogenously regulated CD40L, and the potential for its use in T-cell therapy for X-HIGM syndrome.

Figures