Frontiers in Cell and Developmental Biology (Feb 2024)

Generation of heterozygous and homozygous NF1 lines from human-induced pluripotent stem cells using CRISPR/Cas9 to investigate bone defects associated with neurofibromatosis type 1

  • Annabelle Darle,
  • Thibault Mahiet,
  • Déborah Aubin,
  • Déborah Aubin,
  • Déborah Aubin,
  • Manon Doyen,
  • Manon Doyen,
  • Lina El Kassar,
  • Béatrice Parfait,
  • Béatrice Parfait,
  • Gilles Lemaitre,
  • Gilles Lemaitre,
  • Christine Baldeschi,
  • Christine Baldeschi,
  • Jennifer Allouche,
  • Jennifer Allouche,
  • Nathalie Holic,
  • Nathalie Holic

DOI
https://doi.org/10.3389/fcell.2024.1359561
Journal volume & issue
Vol. 12

Abstract

Read online

Neurofibromatosis type 1 (NF1) is one of the most common genetic disorders caused by heterozygous germline NF1 mutations. NF1 affects many systems, including the skeletal system. To date, no curative therapies are available for skeletal manifestations such as scoliosis and tibial dysplasia, mainly due to the lack of knowledge about the mechanisms that underlie this process. By using CRISPR/Cas9-mediated gene editing in human-induced pluripotent stem cells (hiPSCs) to minimize the variability due to genetic background and epigenetic factors, we generated isogenic heterozygous and homozygous NF1-deficient hiPSC lines to investigate the consequences of neurofibromin inactivation on osteoblastic differentiation. Here, we demonstrate that loss of one or both copies of NF1 does not alter the potential of isogenic hiPSCs to differentiate into mesenchymal stem cells (hiPSC-MSCs). However, NF1 (+/−) and NF1 (−/−) hiPSC-MSCs show a defect in osteogenic differentiation and mineralization. In addition, we show that a mono-allelic deletion in NF1 in an isogenic context is sufficient to impair cell differentiation into osteoblasts. Overall, this study highlights the relevance of generating isogenic lines, which may help in genotype–phenotype correlation and provide a human cellular model to understand the molecular mechanisms underlying NF1 and, thus, discover new therapeutic strategies.

Keywords