4.8 Article

Cellular senescence checkpoint function determines differential Notch1-dependent oncogenic and tumor-suppressor activities

Journal

ONCOGENE
Volume 34, Issue 18, Pages 2347-2359

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/onc.2014.169

Keywords

-

Funding

  1. NIH [P01CA098101, U01CA143056, R01DK077005, K26 RR032714]
  2. Pennsylvania CURE Program Grant [F32-CA174176, F32-DE024685, K08DE022842]
  3. Trio/ACS Career Award
  4. VA CPPF Grant [K07CA137140]
  5. University of Pennsylvania University Research Foundation Award
  6. University of Pennsylvania
  7. Abramson Cancer Center Pilot Project Grant
  8. American Cancer Society [RP-10-033-01-CCE]
  9. Grants-in-Aid for Scientific Research [25460926] Funding Source: KAKEN

Ask authors/readers for more resources

Notch activity regulates tumor biology in a context-dependent and complex manner. Notch may act as an oncogene or a tumor-suppressor gene even within the same tumor type. Recently, Notch signaling has been implicated in cellular senescence. Yet, it remains unclear as to how cellular senescence checkpoint functions may interact with Notch-mediated oncogenic and tumor-suppressor activities. Herein, we used genetically engineered human esophageal keratinocytes and esophageal squamous cell carcinoma cells to delineate the functional consequences of Notch activation and inhibition along with pharmacological intervention and RNA interference experiments. When expressed in a tetracycline-inducible manner, the ectopically expressed activated form of Notch1 (ICN1) displayed oncogene-like characteristics inducing cellular senescence corroborated by the induction of G0/G1 cell-cycle arrest, Rb dephosphorylation, flat and enlarged cell morphology and senescence-associated beta-galactosidase activity. Notch-induced senescence involves canonical CSL/RBPJ-dependent transcriptional activity and the p16(INK4A)-Rb pathway. Loss of p16(INK4A) or the presence of human papilloma virus (HPV) E6/E7 oncogene products not only prevented ICN1 from inducing senescence but permitted ICN1 to facilitate anchorage-independent colony formation and xenograft tumor growth with increased cell proliferation and reduced squamous-cell differentiation. Moreover, Notch1 appears to mediate replicative senescence as well as transforming growth factor-beta-induced cellular senescence in non-transformed cells and that HPV E6/E7 targets Notch1 for inactivation to prevent senescence, revealing a tumor-suppressor attribute of endogenous Notch1. In aggregate, cellular senescence checkpoint functions may influence dichotomous Notch activities in the neoplastic context.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available