1. Academic Validation
  2. UBC9 deficiency enhances immunostimulatory macrophage activation and subsequent antitumor T cell response in prostate cancer

UBC9 deficiency enhances immunostimulatory macrophage activation and subsequent antitumor T cell response in prostate cancer

  • J Clin Invest. 2023 Jan 10;e158352. doi: 10.1172/JCI158352.
Jun Xiao 1 Fei Sun 2 Ya-Nan Wang 1 Bo Liu 3 Peng Zhou 1 Fa-Xi Wang 2 Hai-Feng Zhou 4 Yue Ge 1 Tian-Tian Yue 2 Jia-Hui Luo 2 Chun-Liang Yang 2 Shan-Jie Rong 2 Ze-Zhong Xiong 1 Sheng Ma 1 Qi Zhang 5 Yang Xun 1 Chun-Guang Yang 1 Yang Luan 1 Shao-Gang Wang 1 Cong-Yi Wang 2 Zhi-Hua Wang 1
Affiliations

Affiliations

  • 1 Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
  • 2 Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
  • 3 Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
  • 4 Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
  • 5 Department of Critical Care Medicine, Zhongnan Hospital,Wuhan University, Wuhan, China.
Abstract

The role of tumor-associated macrophages (TAMs) along with the regulatory mechanisms underlying distinct macrophage activation states remain poorly understood in prostate Cancer (PCa). Herein, we reported that PCa growth in macrophage-specific Ubc9 deficient mice is substantially suppressed compared to their wild-type littermates, an effect partially ascribed to the augmented CD8+ T cell response. Biochemical and molecular analyses revealed that the signal transducer and activator of transcription 4 (STAT4) is a crucial UBC9-mediated SUMOylation target, with lysine residue 350 (K350) as the major modification site. Site-directed mutation of STAT4 (K350R) enhanced its nuclear translocation and stability, thereby facilitating the proinflammatory activation of macrophages. Importantly, administration of UBC9 inhibitor, 2-D08, promoted the antitumor effect of TAMs and increased the expression of PD-1 on CD8+ T cells, supporting a synergistic antitumor efficacy once it combined with the immune checkpoint blockade (ICB) therapy. Together, our results demonstrated that ablation of UBC9 could reverse the immunosuppressive phenotype of TAMs via promoting STAT4 mediated macrophage activation and macrophage-CD8+ T cell crosstalk, which provides valuable insights to halt the pathogenic process of tumorigenesis.

Keywords

Cancer immunotherapy; Immunology; Macrophages; Oncology; Prostate cancer.

Figures
Products