Med23 deficiency reprograms the tumor microenvironment to promote lung tumorigenesis

Br J Cancer. 2024 Mar;130(5):716-727. doi: 10.1038/s41416-023-02556-9. Epub 2024 Jan 9.

Abstract

Background: Lung cancer is the leading cause of cancer-related death worldwide. We previously found that Mediator complex subunit 23 (MED23) is important for the tumourigenicity of lung cancer cells with hyperactive Ras activity in vitro, although the in vivo function of MED23 in lung tumourigenesis remains to be explored.

Methods: In this study, we utilized well-characterized KrasG12D-driven non-small cell lung cancer mouse model to investigate the role of MED23 in lung cancer. The lung tumour progression was evaluated by H&E and IHC analysis. Western blotting and qRT-PCR assays were performed to detect changes in gene expression. Immune cells were analyzed by FACS technology. RNA-seq and reporter assays were conducted to explore the mechanism.

Results: We observed that lung epithelial Med23 deletion by adeno-Cre resulted in a significant increase in KrasG12D tumour number and size, which was further verified with another mouse model with Med23 specifically deleted in alveolar type II cells. Mice with lung-specific Med23 deficiency also exhibited accelerated tumourigenesis, and a higher proliferation rate for tumour cells, along with increased ERK phosphorylation. Notably, the numbers of infiltrating CD4+ T cells and CD8+ T cells were significantly reduced in the lungs of Med23-deficient mice, while the numbers of myeloid-derived suppressor cells (MDSCs) and Treg cells were significantly increased, suggesting the enhanced immune escape capability of the Med23-deficient lung tumours. Transcriptomic analysis revealed that the downregulated genes in Med23-deficient lung tumour tissues were associated with the immune response. Specifically, Med23 deficiency may compromise the MHC-I complex formation, partially through down-regulating B2m expression.

Conclusions: Collectively, these findings revealed that MED23 may negatively regulate Kras-induced lung tumourigenesis in vivo, which would improve the precise classification of KRAS-mutant lung cancer patients and provide new insights for clinical interventions.

MeSH terms

  • Animals
  • CD8-Positive T-Lymphocytes / metabolism
  • Carcinogenesis / genetics
  • Carcinoma, Non-Small-Cell Lung* / genetics
  • Cell Transformation, Neoplastic / genetics
  • Humans
  • Lung / metabolism
  • Lung Neoplasms* / pathology
  • Mediator Complex / genetics
  • Mice
  • Proto-Oncogene Proteins p21(ras) / metabolism
  • Tumor Microenvironment / genetics

Substances

  • Proto-Oncogene Proteins p21(ras)
  • Mediator Complex
  • Med23 protein, mouse