Advanced Science (Sep 2024)

M6A‐mediated upregulation of lncRNA TUG1 in liver cancer cells regulates the antitumor response of CD8+ T cells and phagocytosis of macrophages

  • Qing Xi,
  • Guangze Yang,
  • Xue He,
  • Hao Zhuang,
  • Li Li,
  • Bing Lin,
  • Lingling Wang,
  • Xianyang Wang,
  • Chunqiang Fang,
  • Qiurui Chen,
  • Yongjie Yang,
  • Zhaoan Yu,
  • Hao Zhang,
  • Wenqian Cai,
  • Yan Li,
  • Han Shen,
  • Li Liu,
  • Rongxin Zhang

DOI
https://doi.org/10.1002/advs.202400695
Journal volume & issue
Vol. 11, no. 34
pp. n/a – n/a

Abstract

Read online

Abstract Tumor immune evasion relies on the crosstalk between tumor cells and adaptive/innate immune cells. Immune checkpoints play critical roles in the crosstalk, and immune checkpoint inhibitors have achieved promising clinical effects. The long non‐coding RNA taurine‐upregulated gene 1 (TUG1) is upregulated in hepatocellular carcinoma (HCC). However, how TUG1 is upregulated and the effects on tumor immune evasion are incompletely understood. Here, METTL3‐mediated m6A modification led to TUG1 upregulation is demonstrated. Knockdown of TUG1 inhibited tumor growth and metastasis, increased the infiltration of CD8+ T cells and M1‐like macrophages in tumors, promoted the activation of CD8+ T cells through PD‐L1, and improved the phagocytosis of macrophages through CD47. Mechanistically, TUG1 regulated PD‐L1 and CD47 expressions by acting as a sponge of miR‐141 and miR‐340, respectively. Meanwhile, TUG1 interacted with YBX1 to facilitate the upregulation of PD‐L1 and CD47 transcriptionally, which ultimately regulated tumor immune evasion. Clinically, TUG1 positively correlated with PD‐L1 and CD47 in HCC tissues. Moreover, the combination of Tug1‐siRNA therapy with a Pdl1 antibody effectively suppressed tumor growth. Therefore, the mechanism of TUG1 in regulating tumor immune evasion is revealed and can inform existing strategies targeting TUG1 for enhancing HCC immune therapy and drug development.

Keywords