Chromosome silencing in vitro reveals trisomy 21 causes cell-autonomous deficits in angiogenesis and early dysregulation in Notch signaling

Cell Rep. 2022 Aug 9;40(6):111174. doi: 10.1016/j.celrep.2022.111174.

Abstract

Despite the prevalence of Down syndrome (DS), little is known regarding the specific cell pathologies that underlie this multi-system disorder. To understand which cell types and pathways are more directly affected by trisomy 21 (T21), we used an inducible-XIST system to silence one chromosome 21 in vitro. T21 caused the dysregulation of Notch signaling in iPSCs, potentially affecting cell-type programming. Further analyses identified dysregulation of pathways important for two cell types: neurogenesis and angiogenesis. Angiogenesis is essential to many bodily systems, yet is understudied in DS; therefore, we focused next on whether T21 affects endothelial cells. An in vitro assay for microvasculature formation revealed a cellular pathology involving delayed tube formation in response to angiogenic signals. Parallel transcriptomic analysis of endothelia further showed deficits in angiogenesis regulators. Results indicate a direct cell-autonomous impact of T21 on endothelial function, highlighting the importance of angiogenesis, with wide-reaching implications for development and disease progression.

Keywords: CP: Developmental biology; Down syndrome; NOTCH signaling; XIST; angiogenesis; cardiovascular; chromosome silencing; endothelial cell differentiation; human pluripotent stem cells.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Chromosomes / metabolism
  • Down Syndrome* / genetics
  • Down Syndrome* / pathology
  • Endothelial Cells / metabolism
  • Humans
  • Induced Pluripotent Stem Cells* / metabolism
  • Trisomy