Advanced Science (Aug 2024)

CD147‐K148me2‐Driven Tumor Cell‐Macrophage Crosstalk Provokes NSCLC Immunosuppression via the CCL5/CCR5 Axis

  • Ke Wang,
  • Xiaohong Chen,
  • Peng Lin,
  • Jiao Wu,
  • Qiang Huang,
  • Zhi‐Nan Chen,
  • Jiale Tian,
  • Hao Wang,
  • Ye Tian,
  • Mingyan Shi,
  • Meirui Qian,
  • Bengang Hui,
  • Yumeng Zhu,
  • Ling Li,
  • Rui Yao,
  • Huijie Bian,
  • Ping Zhu,
  • Ruo Chen,
  • Liang Chen

DOI
https://doi.org/10.1002/advs.202400611
Journal volume & issue
Vol. 11, no. 29
pp. n/a – n/a

Abstract

Read online

Abstract Immunosuppression is a major hallmark of tumor progression in non‐small cell lung cancer (NSCLC). Cluster of differentiation 147 (CD147), an important pro‐tumorigenic factor, is closely linked to NSCLC immunosuppression. However, the role of CD147 di‐methylation in the immunosuppressive tumor microenvironment (TME) remains unclear. Here, di‐methylation of CD147 at Lys148 (CD147‐K148me2) is identified as a common post‐translational modification (PTM) in NSCLC that is significantly associated with unsatisfying survival outcomes among NSCLC sufferers, especially those in the advanced stages of the disease. The methyltransferase NSD2 catalyzes CD147 to generate CD147‐K148me2. Further analysis demonstrates that CD147‐K148me2 reestablishes the immunosuppressive TME and promotes NSCLC progression. Mechanistically, this modification promotes the interaction between cyclophilin A (CyPA) and CD147, and in turn, increases CCL5 gene transcription by activating p38‐ZBTB32 signaling, leading to increased NSCLC cell‐derived CCL5 secretion. Subsequently, CD147‐K148me2‐mediated CCL5 upregulation facilitates M2‐like tumor‐associated macrophage (TAM) infiltration in NSCLC tissues via CCL5/CCR5 axis‐dependent intercellular crosstalk between tumor cells and macrophages, which is inhibited by blocking CD147‐K148me2 with the targeted antibody 12C8. Overall, this study reveals the role of CD147‐K148me2‐driven intercellular crosstalk in the development of NSCLC immunosuppression, and provides a potential interventional strategy for PTM‐targeted NSCLC therapy.

Keywords