A stress-induced cilium-to-PML-NB route drives senescence initiation

Nat Commun. 2023 Apr 3;14(1):1840. doi: 10.1038/s41467-023-37362-7.

Abstract

Cellular senescence contributes to tissue homeostasis and age-related pathologies. However, how senescence is initiated in stressed cells remains vague. Here, we discover that exposure to irradiation, oxidative or inflammatory stressors induces transient biogenesis of primary cilia, which are then used by stressed cells to communicate with the promyelocytic leukemia nuclear bodies (PML-NBs) to initiate senescence responses in human cells. Mechanistically, a ciliary ARL13B-ARL3 GTPase cascade negatively regulates the association of transition fiber protein FBF1 and SUMO-conjugating enzyme UBC9. Irreparable stresses downregulate the ciliary ARLs and release UBC9 to SUMOylate FBF1 at the ciliary base. SUMOylated FBF1 then translocates to PML-NBs to promote PML-NB biogenesis and PML-NB-dependent senescence initiation. Remarkably, Fbf1 ablation effectively subdues global senescence burden and prevents associated health decline in irradiation-treated mice. Collectively, our findings assign the primary cilium a key role in senescence induction in mammalian cells and, also, a promising target in future senotherapy strategies.

Publication types

  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, Non-P.H.S.
  • Research Support, N.I.H., Extramural

MeSH terms

  • Adaptor Proteins, Signal Transducing / metabolism
  • Animals
  • Cell Nucleus / metabolism
  • Cilia* / metabolism
  • Humans
  • Mammals / metabolism
  • Mice
  • Nuclear Proteins* / metabolism
  • Promyelocytic Leukemia Nuclear Bodies
  • Promyelocytic Leukemia Protein / metabolism
  • Sumoylation

Substances

  • Promyelocytic Leukemia Protein
  • Nuclear Proteins
  • Fbf1 protein, mouse
  • Adaptor Proteins, Signal Transducing