Abstract

Abstract The classification of patient tumors by clinical subtype has gained wide acceptance due to the implications for prognosis and treatment. However, recent studies have cast doubt on previously advocated normal mammary origins of these subtypes. Thus, the link between the normal mammary gland and mammary tumors is more complex than expected. C-Jun N-Terminal Kinase-2 (JNK2) is a protein that is involved in numerous developmental processes and our previous work has shown it to be important for DNA damage response in mammary tumors. In attempt to gain insight into the link between mammary development and tumorigenesis, we compared normal mammary glands of JNK2 knockout (jnk2ko) mice to jnk2ko mammary tumors expressing or lacking wildtype p53 (p53ko). These studies showed that jnk2ko glands possess 35% fewer basal cells (p=0.0078) with a corresponding increase in luminal epithelial cell populations (p=0.100). This luminal response is corroborated by in vitro 3D assays of primary mammary epithelial cells (MECs) where luminal cell differentiation is normalized by inhibition of Notch signaling. Expression notch-1, a well-known regulator of MEC differentiation, is increased jnk2ko mammary glands. Increased expression of the Notch-1 target gene, hes-1, was also seen (p=0.005). Histology revealed that increased expression of active Notch-1 is localized to the mammary stem cell niche, the terminal end bud. Similar to the normal gland, jnk2ko mammary tumors possessing wildtype p53 exhibit decreased proportions of basal cells (p=0.0002) and increased proportions of luminal cells (p=0.0411) relative to wildtype. Jnk2ko cell lines derived from these tumors show decreased expression of notch-1 (p=0.0018) and hes-1 (p=0.0602) following introduction of JNK2. Luciferase assays comparing activity of the notch-1 promoter to a notch-1 promoter with mutated p53 response elements revealed a dependence of increased notch-1 promoter activity in jnk2ko cells on the p53 response element. P53ko tumor cells, by contrast, do not exhibit alterations in notch-1 promoter activity in the absence of p53 response elements, regardless of JNK2 status. QPCR showed that loss of JNK2 in normal mammary glands and tumors causes increased p53 expression—thus providing a potential mechanism. In support that Notch upregulation in the absence of JNK2 is dependent upon p53, normal glands lacking p53 show no differences in lineage differentiation. P53ko tumors also show no differences in basal lineage differentiation, however, increases in luminal differentiation are maintained in the absence of JNK2. Consistent with increased luminal differentiation, jnk2ko caused decreased expression of markers involved in the epithelial to mesenchymal transition phenotype. This data suggests that JNK2 is important not only for lineage differentiation in normal mammary glands, but in mammary tumors and that the effect is dependent on both Notch1 and p53. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr PD08-01.

Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.