Correlation Engine 2.0
Clear Search sequence regions


  • cancer (9)
  • CD163 (1)
  • CEBPB (1)
  • chromatin (2)
  • cofactors (1)
  • cytokines (1)
  • factor (2)
  • humans (1)
  • il 4 (1)
  • IL 6 (7)
  • Interleukin 6 (2)
  • lung metastasis (1)
  • lymph node (1)
  • macrophage (13)
  • mice (4)
  • patients (1)
  • PD 1 (1)
  • phase (3)
  • prostate (11)
  • protein human (1)
  • rna (1)
  • signals (1)
  • STAT6 (1)
  • t lymphocytes (1)
  • YY1 (14)
  • yy1 protein, human (1)
  • Sizes of these terms reflect their relevance to your search.

    Tumor-associated macrophages are mainly polarized into the M2 phenotype, remodeling the tumor microenvironment and promoting tumor progression by secreting various cytokines. Tissue microarray consisting of prostate cancer (PCa), normal prostate, and lymph node metastatic samples from patients with PCa were stained with Yin Yang 1 (YY1) and CD163. Transgenic mice overexpressing YY1 were constructed to observe PCa tumorigenesis. Furthermore, in vivo and in vitro experiments, including CRISPR-Cas9 knock-out, RNA sequencing, chromatin immunoprecipitation (ChIP) sequencing, and liquid-liquid phase separation (LLPS) assays, were performed to investigate the role and mechanism of YY1 in M2 macrophages and PCa tumor microenvironment. YY1 was highly expressed in M2 macrophages in PCa and was associated with poorer clinical outcomes. The proportion of tumor-infiltrated M2 macrophages increased in transgenic mice overexpressing YY1. In contrast, the proliferation and activity of anti-tumoral T lymphocytes were suppressed. Treatment targeting YY1 on M2 macrophages using an M2-targeting peptide-modified liposome carrier suppressed PCa cell lung metastasis and generated synergistic anti-tumoral effects with PD-1 blockade. IL-4/STAT6 pathway regulated YY1, and YY1 increased the macrophage-induced PCa progression by upregulating IL-6. Furthermore, by conducting H3K27ac-ChIP-seq in M2 macrophages and THP-1, we found that thousands of enhancers were gained during M2 macrophage polarization, and these M2-specific enhancers were enriched in YY1 ChIP-seq signals. In addition, an M2-specific IL-6 enhancer upregulated IL-6 expression through long-range chromatin interaction with IL-6 promoter in M2 macrophages. During M2 macrophage polarization, YY1 formed an LLPS, in which p300, p65, and CEBPB acted as transcriptional cofactors. Phase separation of the YY1 complex in M2 macrophages upregulated IL-6 by promoting IL-6 enhancer-promoter interactions, thereby increasing PCa progression. © Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

    Citation

    Saisai Chen, Kai Lu, Yue Hou, Zonghao You, Chuanjun Shu, Xiaoying Wei, Tiange Wu, Naipeng Shi, Guangyuan Zhang, Jianping Wu, Shuqiu Chen, Lihua Zhang, Wenchao Li, Dingxiao Zhang, Shenghong Ju, Ming Chen, Bin Xu. YY1 complex in M2 macrophage promotes prostate cancer progression by upregulating IL-6. Journal for immunotherapy of cancer. 2023 Apr;11(4)

    Expand section icon Mesh Tags

    Expand section icon Substances


    PMID: 37094986

    View Full Text