Abstract

Cigarette smoking is undoubtedly a risk factor for lung cancer. Moreover, smokers with genetic mutations on chromosome 3p21.3, a region frequently deleted in cancer and notably in lung cancer, have a dramatically higher risk of aggressive lung cancer. The RNA binding motif 5 (RBM5) is one of the component genes in the 3p21.3 tumour suppressor region. Studies using human cancer specimens and cell lines suggest a role for RBM5 as a tumour suppressor. Here we demonstrate, for the first time, an in vivo role for RBM5 as a tumour suppressor in the mouse lung. We generated Rbm5 loss-of-function mice and exposed them to a tobacco carcinogen NNK. Upon exposure to NNK, Rbm5 loss-of-function mice developed lung cancer at similar rates to wild type mice. As tumourigenesis progressed, however, reduced Rbm5 expression lead to significantly more aggressive lung cancer i.e. increased adenocarcinoma nodule numbers and tumour size. Our data provide in vivo evidence that reduced RBM5 function, as occurs in a large number of patients, coupled with exposure to tobacco carcinogens is a risk factor for an aggressive lung cancer phenotype. These data suggest that RBM5 loss-of-function likely underpins at least part of the pro-tumourigenic consequences of 3p21.3 deletion in humans.

Highlights

  • Late stage detection makes lung cancer one of the most fatal forms of cancer, with a five-year survival rate of 17% overall, or below 2% for those with stage IV disease at diagnosis[1]

  • RNA binding motif 5 (RBM5) is one of the genes located within the tumour suppressor region 3p21.3; a region containing 19 genes that is frequently deleted in lung cancer and other types of carcinomas[3]

  • Human lung cancers can be divided into two main histopathological subtypes: non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC)

Read more

Summary

Results and Discussion

To test the in vivo function of RBM5, we generated an Rbm[5] knockout mouse line using a gene trapped ES cell line. In this study, we utilized NNK to induce lung cancer in Rbm[5] heterozygous knockout (carrying one copy of the Rbm[5] gene) and wild type mice. Blinded, analysis by a pathologist confirmed that the Rbm5+/− lungs contained an increased numbers of adenocarcinoma nodules with increased tumour nodule sizes compared to that in Rbm5+/+ lungs (148% and 153% increase on control wild type tissue respectively) (Fig. 3, Supplementary Table 1) These data indicate that reduced Rbm[5] expression leads to more aggressive progression of lung adenocarcinomas in the Rbm5+/− mice. Consistent with previous publications suggesting at AEC cells are the origin of many lung cancers[20], tumours in both Rbm5+/+ and Rbm5+/− mice were Pro-SPC positive, and CC10 negative (Supplementary Fig. 3). Our data reveal that RBM5 is required for early post-natal survival and acts in vivo as a tumour suppressor that likely underpins at least part of the pro-tumourigenic outcomes resulting from 3p21.3 deletion in humans

Methods
Author Contributions
Additional Information
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call