Cell Death and Disease (Aug 2023)

CRISPR screen identifies the role of RBBP8 in mediating unfolded protein response induced liver damage through regulating protein synthesis

  • Heting Wang,
  • Xuya Pan,
  • Xiaoxin Xiang,
  • Yang Zhang,
  • Jianning Chen,
  • Shiyi Wen,
  • Jin Wang,
  • Rong Gao,
  • Jifeng Yang,
  • Yaping Zhi,
  • Siying Wen,
  • Yubao Zheng,
  • Ting Li,
  • Heying Ai,
  • Xuemin He,
  • Yan Lu,
  • Yanhua Zhu,
  • Chunliang Li,
  • Yanming Chen,
  • Guojun Shi

DOI
https://doi.org/10.1038/s41419-023-06046-x
Journal volume & issue
Vol. 14, no. 8
pp. 1 – 12

Abstract

Read online

Abstract Unfolded protein response (UPR) maintains the endoplasmic reticulum (ER) homeostasis, survival, and physiological function of mammalian cells. However, how cells adapt to ER stress under physiological or disease settings remains largely unclear. Here by a genome-wide CRISPR screen, we identified that RBBP8, an endonuclease involved in DNA damage repair, is required for ATF4 activation under ER stress in vitro. RNA-seq analysis suggested that RBBP8 deletion led to impaired cell cycle progression, retarded proliferation, attenuated ATF4 activation, and reduced global protein synthesis under ER stress. Mouse tissue analysis revealed that RBBP8 was highly expressed in the liver, and its expression is responsive to ER stress by tunicamycin intraperitoneal injection. Hepatocytes with RBBP8 inhibition by adenovirus-mediated shRNA were resistant to tunicamycin (Tm)-induced liver damage, cell death, and ER stress response. To study the pathological role of RBBP8 in regulating ATF4 activity, we illustrated that both RBBP8 and ATF4 were highly expressed in liver cancer tissues compared with healthy controls and highly expressed in Ki67-positive proliferating cells within the tumors. Interestingly, overexpression of RBBP8 in vitro promoted ATF4 activation under ER stress, and RBBP8 expression showed a positive correlation with ATF4 expression in liver cancer tissues by co-immunostaining. Our findings provide new insights into the mechanism of how cells adapt to ER stress through the crosstalk between the nucleus and ER and how tumor cells survive under chemotherapy or other anticancer treatments, which suggests potential therapeutic strategies against liver disease by targeting DNA damage repair, UPR or protein synthesis.