[HTML][HTML] N-cadherin upregulation mediates adaptive radioresistance in glioblastoma

S Osuka, D Zhu, Z Zhang, C Li… - The Journal of …, 2021 - Am Soc Clin Investig
S Osuka, D Zhu, Z Zhang, C Li, CT Stackhouse, O Sampetrean, JJ Olson, GY Gillespie
The Journal of clinical investigation, 2021Am Soc Clin Investig
Glioblastoma (GBM) is composed of heterogeneous tumor cell populations, including those
with stem cell properties, termed glioma stem cells (GSCs). GSCs are innately less radiation
sensitive than the tumor bulk and are believed to drive GBM formation and recurrence after
repeated irradiation. However, it is unclear how GSCs adapt to escape the toxicity of
repeated irradiation used in clinical practice. To identify important mediators of adaptive
radioresistance in GBM, we generated radioresistant human and mouse GSCs by exposing …
Glioblastoma (GBM) is composed of heterogeneous tumor cell populations, including those with stem cell properties, termed glioma stem cells (GSCs). GSCs are innately less radiation sensitive than the tumor bulk and are believed to drive GBM formation and recurrence after repeated irradiation. However, it is unclear how GSCs adapt to escape the toxicity of repeated irradiation used in clinical practice. To identify important mediators of adaptive radioresistance in GBM, we generated radioresistant human and mouse GSCs by exposing them to repeat cycles of irradiation. Surviving subpopulations acquired strong radioresistance in vivo, which was accompanied by a reduction in cell proliferation and an increase in cell-cell adhesion and N-cadherin expression. Increasing N-cadherin expression rendered parental GSCs radioresistant, reduced their proliferation, and increased their stemness and intercellular adhesive properties. Conversely, radioresistant GSCs lost their acquired phenotypes upon CRISPR/Cas9-mediated knockout of N-cadherin. Mechanistically, elevated N-cadherin expression resulted in the accumulation of β-catenin at the cell surface, which suppressed Wnt/β-catenin proliferative signaling, reduced neural differentiation, and protected against apoptosis through Clusterin secretion. N-cadherin upregulation was induced by radiation-induced IGF1 secretion, and the radiation resistance phenotype could be reverted with picropodophyllin, a clinically applicable blood-brain-barrier permeable IGF1 receptor inhibitor, supporting clinical translation.
The Journal of Clinical Investigation