Frontiers in Immunology (Jun 2021)

Premature Senescence and Increased Oxidative Stress in the Thymus of Down Syndrome Patients

  • Genni Enza Marcovecchio,
  • Francesca Ferrua,
  • Francesca Ferrua,
  • Francesca Ferrua,
  • Elena Fontana,
  • Elena Fontana,
  • Stefano Beretta,
  • Marco Genua,
  • Ileana Bortolomai,
  • Anastasia Conti,
  • Davide Montin,
  • Davide Montin,
  • Maria Teresa Cascarano,
  • Sonia Bergante,
  • Veronica D’Oria,
  • Alessandro Giamberti,
  • Donato Amodio,
  • Donato Amodio,
  • Caterina Cancrini,
  • Caterina Cancrini,
  • Adriano Carotti,
  • Raffaella Di Micco,
  • Ivan Merelli,
  • Marita Bosticardo,
  • Marita Bosticardo,
  • Anna Villa,
  • Anna Villa

DOI
https://doi.org/10.3389/fimmu.2021.669893
Journal volume & issue
Vol. 12

Abstract

Read online

Down syndrome (DS) patients prematurely show clinical manifestations usually associated with aging. Their immune system declines earlier than healthy individuals, leading to increased susceptibility to infections and higher incidence of autoimmune phenomena. Clinical features of accelerated aging indicate that trisomy 21 increases the biological age of tissues. Based on previous studies suggesting immune senescence in DS, we hypothesized that induction of cellular senescence may contribute to early thymic involution and immune dysregulation. Immunohistochemical analysis of thymic tissue showed signs of accelerated thymic aging in DS patients, normally seen in older healthy subjects. Moreover, our whole transcriptomic analysis on human Epcam-enriched thymic epithelial cells (hTEC), isolated from three DS children, which revealed disease-specific transcriptomic alterations. Gene set enrichment analysis (GSEA) of DS TEC revealed an enrichment in genes involved in cellular response to stress, epigenetic histone DNA modifications and senescence. Analysis of senescent markers and oxidative stress in hTEC and thymocytes confirmed these findings. We detected senescence features in DS TEC, thymocytes and peripheral T cells, such as increased β-galactosidase activity, increased levels of the cell cycle inhibitor p16, telomere length and integrity markers and increased levels of reactive oxygen species (ROS), all factors contributing to cellular damage. In conclusion, our findings support the key role of cellular senescence in the pathogenesis of immune defect in DS while adding new players, such as epigenetic regulation and increased oxidative stress, to the pathogenesis of immune dysregulation.

Keywords