Your browser doesn't support javascript.
loading
Cytosolic DNA sensor AIM2 promotes KRAS-driven lung cancer independent of inflammasomes.
Alanazi, Mohammad; Weng, Teresa; McLeod, Louise; Gearing, Linden J; Smith, Julian A; Kumar, Beena; Saad, Mohamed I; Jenkins, Brendan J.
Afiliación
  • Alanazi M; Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
  • Weng T; Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia.
  • McLeod L; Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
  • Gearing LJ; Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia.
  • Smith JA; Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
  • Kumar B; Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia.
  • Saad MI; Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
  • Jenkins BJ; Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia.
Cancer Sci ; 115(6): 1834-1850, 2024 Jun.
Article en En | MEDLINE | ID: mdl-38594840
ABSTRACT
Constitutively active KRAS mutations are among the major drivers of lung cancer, yet the identity of molecular co-operators of oncogenic KRAS in the lung remains ill-defined. The innate immune cytosolic DNA sensor and pattern recognition receptor (PRR) Absent-in-melanoma 2 (AIM2) is best known for its assembly of multiprotein inflammasome complexes and promoting an inflammatory response. Here, we define a role for AIM2, independent of inflammasomes, in KRAS-addicted lung adenocarcinoma (LAC). In genetically defined and experimentally induced (nicotine-derived nitrosamine ketone; NNK) LAC mouse models harboring the KrasG12D driver mutation, AIM2 was highly upregulated compared with other cytosolic DNA sensors and inflammasome-associated PRRs. Genetic ablation of AIM2 in KrasG12D and NNK-induced LAC mouse models significantly reduced tumor growth, coincident with reduced cellular proliferation in the lung. Bone marrow chimeras suggest a requirement for AIM2 in KrasG12D-driven LAC in both hematopoietic (immune) and non-hematopoietic (epithelial) cellular compartments, which is supported by upregulated AIM2 expression in immune and epithelial cells of mutant KRAS lung tissues. Notably, protection against LAC in AIM2-deficient mice is associated with unaltered protein levels of mature Caspase-1 and IL-1ß inflammasome effectors. Moreover, genetic ablation of the key inflammasome adapter, ASC, did not suppress KrasG12D-driven LAC. In support of these in vivo findings, AIM2, but not mature Caspase-1, was upregulated in human LAC patient tumor biopsies. Collectively, our findings reveal that endogenous AIM2 plays a tumor-promoting role, independent of inflammasomes, in mutant KRAS-addicted LAC, and suggest innate immune DNA sensing may provide an avenue to explore new therapeutic strategies in lung cancer.
Asunto(s)
Palabras clave

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Proteínas Proto-Oncogénicas p21(ras) / Proteínas de Unión al ADN / Inflamasomas / Adenocarcinoma del Pulmón / Neoplasias Pulmonares Idioma: En Revista: Cancer Sci Año: 2024 Tipo del documento: Article

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Proteínas Proto-Oncogénicas p21(ras) / Proteínas de Unión al ADN / Inflamasomas / Adenocarcinoma del Pulmón / Neoplasias Pulmonares Idioma: En Revista: Cancer Sci Año: 2024 Tipo del documento: Article