Login / Signup

Sex specific function of epithelial STAT3 signaling in pathogenesis of K-ras mutant lung cancer.

Mauricio S CaetanoMaya HassaneHieu T VanEmmanuel BugarinAmber M CumpianChristina L McDowellCarolina Gonzalez CavazosHuiyuan ZhangShanshan DengLixia DiaoJing WangScott E EvansCarmen BehrensIgnacio I WistubaSusan A W FuquaHuang LinLaura P StabileStephanie S WatowichHumam KadaraSeyed Javad Moghaddam
Published in: Nature communications (2018)
Lung adenocarcinomas (LUADs) with mutations in the K-ras oncogene display dismal prognosis. Proinflammatory and immunomodulatory events that drive development of K-ras mutant LUAD are poorly understood. Here, we develop a lung epithelial specific K-ras mutant/Stat3 conditional knockout (LR/Stat3Δ/Δ) mouse model. Epithelial Stat3 deletion results in intriguing sex-associated discrepancies; K-ras mutant tumors are decreased in female LR/Stat3Δ/Δ mice whereas tumor burdens are increased in males. RNA-sequencing and tumor microenvironment (TME) analysis demonstrate increased anti-tumor immune responses following Stat3 deletion in females and, conversely, elevated pro-tumor immune pathways in males. While IL-6 blockade in male LR/Stat3Δ/Δ mice reduces lung tumorigenesis, inhibition of estrogen receptor signaling in female mice augments K-ras mutant oncogenesis and reprograms lung TME toward a pro-tumor phenotype. Our data underscore a critical sex-specific role for epithelial Stat3 signaling in K-ras mutant LUAD, thus paving the way for developing personalized (e.g. sex-based) immunotherapeutic strategies for this fatal disease.
Keyphrases
  • wild type
  • cell proliferation
  • immune response
  • mouse model
  • type diabetes
  • anti inflammatory
  • dendritic cells
  • big data
  • artificial intelligence
  • data analysis
  • deep learning