Abstract

Notch signaling plays a complex role in carcinogenesis, and its signaling pathway has both tumor suppressor and oncogenic components. To identify regulators that might control this dual activity of NOTCH1, we screened a chemical library targeting kinases and identified Polo-like kinase 1 (PLK1) as one of the kinases involved in arsenite-induced NOTCH1 down-modulation. As PLK1 activity drives mitotic entry but also is inhibited after DNA damage, we investigated the PLK1-NOTCH1 interplay in the G2 phase of the cell cycle and in response to DNA damage. Here, we found that PLK1 regulates NOTCH1 expression at G2/M transition. However, when cells in G2 phase are challenged with DNA damage, PLK1 is inhibited to prevent entry into mitosis. Interestingly, we found that the interaction between NOTCH1 and PLK1 is functionally important during the DNA damage response, as we found that whereas PLK1 activity is inhibited, NOTCH1 expression is maintained during DNA damage response. During genotoxic stress, cellular transformation requires that promitotic activity must override DNA damage checkpoint signaling to drive proliferation. Interestingly, we found that arsenite-induced genotoxic stress causes a PLK1-dependent signaling response that antagonizes the involvement of NOTCH1 in the DNA damage checkpoint. Taken together, our data provide evidence that Notch signaling is altered but not abolished in SCC cells. Thus, it is also important to recognize that Notch plasticity might be modulated and could represent a key determinant to switch on/off either the oncogenic or tumor suppressor function of Notch signaling in a single type of tumor.

Highlights

  • Notch signaling plays a complex role in carcinogenesis, and its signaling pathway has both tumor suppressor and oncogenic components

  • We found that the interaction between NOTCH1 and Polo-like kinase 1 (PLK1) is functionally important during the DNA damage response, as we found that whereas PLK1 activity is inhibited, NOTCH1 expression is maintained during DNA damage response

  • We previously demonstrated that whereas arsenite-mediated apoptosis of immortalized keratinocytes was associated with NOTCH1 down-regulation, arsenite-mediated transformation of these cells was characterized by increased NOTCH1 stability [12]

Read more

Summary

Edited by Patrick Sung

Notch signaling plays a complex role in carcinogenesis, and its signaling pathway has both tumor suppressor and oncogenic components. We found that arsenite-induced genotoxic stress causes a PLK1-dependent signaling response that antagonizes the involvement of NOTCH1 in the DNA damage checkpoint. The molecular regulation of the dichotomous function of NOTCH signaling remains poorly understood For this reason, we studied this dual activity of NOTCH1 in arsenic-induced keratinocyte transformation, providing a model to investigate the molecular aspects determining whether NOTCH signaling will be either oncogenic or tumor-suppressive [12]. To identify regulators that may influence the dichotomous NOTCH1 function, we screened a chemical library targeting human kinases and identified Polo-like kinase 1 (PLK1) as one of the kinases involved in arsenite-induced down-modulation of NOTCH1 expression. Our data show that NOTCH1 has pleiotropic effects in DNA damage-arrested cells, and in those contexts where NOTCH1 is known to play a tumor suppressor function, cancer cells might still be dependent on specific NOTCH1 signals to sustain their cancerous phenotype

Results
Discussion
Cell culture and transfection
Reagents and immunoblotting
Kinase library screening
Cell cycle analysis
Synchronization and recovery from DNA damage
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