Allele-Specific Disruption of a Common STAT3 Autosomal Dominant Allele Is Not Sufficient to Restore Downstream Signaling in Patient-Derived T Cells

Genes (Basel). 2022 Oct 20;13(10):1912. doi: 10.3390/genes13101912.

Abstract

Dominant negative mutations in the STAT3 gene account for autosomal dominant hyper-IgE syndrome (AD-HIES). Patients typically present high IgE serum levels, recurrent infections, and soft tissue abnormalities. While current therapies focus on alleviating the symptoms, hematopoietic stem cell transplantation (HSCT) has recently been proposed as a strategy to treat the immunological defect and stabilize the disease, especially in cases with severe lung infections. However, because of the potentially severe side effects associated with allogeneic HSCT, this has been considered only for a few patients. Autologous HSCT represents a safer alternative but it requires the removal of the dominant negative mutation in the patients' cells prior to transplantation. Here, we developed allele-specific CRISPR-Cas9 nucleases to selectively disrupt five of the most common STAT3 dominant negative alleles. When tested ex vivo in patient-derived hematopoietic cells, allele-specific disruption frequencies varied in an allele-dependent fashion and reached up to 62% of alleles harboring the V637M mutation without detectable alterations in the healthy STAT3 allele. However, assessment of the gene expression profiles of the STAT3 downstream target genes revealed that, upon activation of those edited patient cells, mono-allelic STAT3 expression (functional haploinsufficiency) is not able to sufficiently restore STAT3-dependent signaling in edited T cells cultured in vitro. Moreover, the stochastic mutagenesis induced by the repair of the nuclease-induced DNA break could further contribute to dominant negative effects. In summary, our results advocate for precise genome editing strategies rather than allele-specific gene disruption to correct the underlying mutations in AD-HIES.

Keywords: CRISPR-Cas; allele-specific nuclease; gene therapy; genome editing; primary immunodeficiency.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Alleles
  • Humans
  • Immunoglobulin E* / genetics
  • Job Syndrome* / diagnosis
  • Job Syndrome* / genetics
  • Job Syndrome* / therapy
  • STAT3 Transcription Factor / genetics
  • STAT3 Transcription Factor / metabolism
  • Signal Transduction / genetics
  • T-Lymphocytes / metabolism

Substances

  • Immunoglobulin E
  • STAT3 protein, human
  • STAT3 Transcription Factor

Grants and funding

This research was funded in part by a sponsored research collaboration with Casebia Therapeutics [to T.C., C.M. and T.I.C.], the Job Research Foundation [to T.C., T.I.C., and B.G.], and internal funding [to C.M.]. B.G. is funded by the Deutsche Forschungsgemeinschaft (SFB1160/2_B5; RESIST–EXC 2155–Project ID 390874280; the EU-H2020-MSCA-COFUND EURIdoc program (No. 101034170) and the BMBF (GAIN 01GM1910A). The article processing charge was funded by the Baden-Wuerttemberg Ministry of Science, Research and Art and the University of Freiburg in the funding program Open Access Publishing.