Signal Transduction and Targeted Therapy (Jun 2024)

Targeting a disintegrin and metalloprotease (ADAM) 17-CD122 axis enhances CD8+ T cell effector differentiation and anti-tumor immunity

  • Lina Sun,
  • Anjun Jiao,
  • Haiyan Liu,
  • Renyi Ding,
  • Ning Yuan,
  • Biao Yang,
  • Cangang Zhang,
  • Xiaoxuan Jia,
  • Gang Wang,
  • Yanhong Su,
  • Dan Zhang,
  • Lin Shi,
  • Chenming Sun,
  • Aijun Zhang,
  • Lianjun Zhang,
  • Baojun Zhang

DOI
https://doi.org/10.1038/s41392-024-01873-6
Journal volume & issue
Vol. 9, no. 1
pp. 1 – 15

Abstract

Read online

Abstract CD8+ T cell immune responses are regulated by multi-layer networks, while the post-translational regulation remains largely unknown. Transmembrane ectodomain shedding is an important post-translational process orchestrating receptor expression and signal transduction through proteolytic cleavage of membrane proteins. Here, by targeting the sheddase A Disintegrin and Metalloprotease (ADAM)17, we defined a post-translational regulatory mechanism mediated by the ectodomain shedding in CD8+ T cells. Transcriptomic and proteomic analysis revealed the involvement of post-translational regulation in CD8+ T cells. T cell-specific deletion of ADAM17 led to a dramatic increase in effector CD8+ T cell differentiation and enhanced cytolytic effects to eliminate pathogens and tumors. Mechanistically, ADAM17 regulated CD8+ T cells through cleavage of membrane CD122. ADAM17 inhibition led to elevated CD122 expression and enhanced response to IL-2 and IL-15 stimulation in both mouse and human CD8+ T cells. Intriguingly, inhibition of ADAM17 in CD8+ T cells improved the efficacy of chimeric antigen receptor (CAR) T cells in solid tumors. Our findings reveal a critical post-translational regulation in CD8+ T cells, providing a potential therapeutic strategy of targeting ADAM17 for effective anti-tumor immunity.