CRISPR screen identifies CEBPB as contributor to dyskeratosis congenita fibroblast senescence via augmented inflammatory gene response

G3 (Bethesda). 2023 Nov 1;13(11):jkad207. doi: 10.1093/g3journal/jkad207.

Abstract

Aging is the consequence of intra- and extracellular events that promote cellular senescence. Dyskeratosis congenita (DC) is an example of a premature aging disorder caused by underlying telomere/telomerase-related mutations. Cells from these patients offer an opportunity to study telomere-related aging and senescence. Our previous work has found that telomere shortening stimulates DNA damage responses (DDRs) and increases reactive oxygen species (ROS), thereby promoting entry into senescence. This work also found that telomere elongation via TERT expression, the catalytic component of the telomere-elongating enzyme telomerase, or p53 shRNA could decrease ROS by disrupting this telomere-DDR-ROS pathway. To further characterize this pathway, we performed a CRISPR/Cas9 knockout screen to identify genes that extend life span in DC cells. Of the cellular clones isolated due to increased life span, 34% had a guide RNA (gRNA) targeting CEBPB, while gRNAs targeting WSB1, MED28, and p73 were observed multiple times. CEBPB is a transcription factor associated with activation of proinflammatory response genes suggesting that inflammation may be present in DC cells. The inflammatory response was investigated using RNA sequencing to compare DC and control cells. Expression of inflammatory genes was found to be significantly elevated (P < 0.0001) in addition to a key subset of these inflammation-related genes [IL1B, IL6, IL8, IL12A, CXCL1 (GROa), CXCL2 (GROb), and CXCL5]. which are regulated by CEBPB. Exogenous TERT expression led to downregulation of RNA/protein CEBPB expression and the inflammatory response genes suggesting a telomere length-dependent mechanism to regulate CEBPB. Furthermore, unlike exogenous TERT and p53 shRNA, CEBPB shRNA did not significantly decrease ROS suggesting that CEBPB's contribution in DC cells' senescence is ROS independent. Our findings demonstrate a key role for CEBPB in engaging senescence by mobilizing an inflammatory response within DC cells.

Keywords: CEBPB; MED28; WSB1; p73; CRISPR; Dyskeratosis congenita; human primary fibroblasts; inflammatory response; oxidative stress; telomere.

Publication types

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

MeSH terms

  • CCAAT-Enhancer-Binding Protein-beta / genetics
  • CCAAT-Enhancer-Binding Protein-beta / metabolism
  • Dyskeratosis Congenita* / genetics
  • Dyskeratosis Congenita* / metabolism
  • Fibroblasts / metabolism
  • Humans
  • Inflammation / genetics
  • Mediator Complex / genetics
  • Mutation
  • RNA, Small Interfering / metabolism
  • Reactive Oxygen Species / metabolism
  • Telomerase* / genetics
  • Telomerase* / metabolism
  • Telomere / genetics
  • Telomere / metabolism
  • Tumor Suppressor Protein p53 / genetics

Substances

  • Reactive Oxygen Species
  • Telomerase
  • Tumor Suppressor Protein p53
  • RNA, Small Interfering
  • MED28 protein, human
  • Mediator Complex
  • CEBPB protein, human
  • CCAAT-Enhancer-Binding Protein-beta