Abstract

Ionizing radiation (IR) principally acts through induction of DNA damage that promotes cell death, although the biological effects of IR are more broad ranging. In fact, the impact of IR of higher-linear energy transfer (LET) on cell biology is generally not well understood. Critically, therefore, the cellular enzymes and mechanisms responsible for enhancing cell survival following high-LET IR are unclear. To this effect, we have recently performed siRNA screening to identify deubiquitylating enzymes that control cell survival specifically in response to high-LET α-particles and protons, in comparison to low-LET X-rays and protons. From this screening, we have now thoroughly validated that depletion of the ubiquitin-specific protease 9X (USP9X) in HeLa and oropharyngeal squamous cell carcinoma (UMSCC74A) cells using small interfering RNA (siRNA), leads to significantly decreased survival of cells after high-LET radiation. We consequently investigated the mechanism through which this occurs, and demonstrate that an absence of USP9X has no impact on DNA damage repair post-irradiation nor on apoptosis, autophagy, or senescence. We discovered that USP9X is required to stabilize key proteins (CEP55 and CEP131) involved in centrosome and cilia formation and plays an important role in controlling pericentrin-rich foci, particularly in response to high-LET protons. This was also confirmed directly by demonstrating that depletion of CEP55/CEP131 led to both enhanced radiosensitivity of cells to high-LET protons and amplification of pericentrin-rich foci. Our evidence supports the importance of USP9X in maintaining centrosome function and biogenesis and which is crucial particularly in the cellular response to high-LET radiation.

Highlights

  • Ionizing radiation (IR) is a major cancer treatment modality for primary and metastatic cancers

  • This led us to identify that depletion of ubiquitin-specific protease 9X (USP9X) caused no reduction in cell survival after low-linear energy transfer (LET) X-ray or proton irradiation, whereas survival after high-LET a-particles and protons was reduced by ~20% and ~40%, respectively (Tables 1, 2)

  • We recently demonstrated using a deubiquitylation enzymes (DUBs) small interfering RNA (siRNA) screen, that depletion of ubiquitin specific protease 6 (USP6) causes significantly increased radiosensitivity to high-LET IR but not low-LET IR, which was mediated by instability of poly (ADPribose) polymerase-1 (PARP-1) required for complex DNA damage (CDD) repair [7]

Read more

Summary

Introduction

Ionizing radiation (IR) is a major cancer treatment modality for primary and metastatic cancers. The biological impact of high-LET radiation is largely associated with the direct effects of irreparable CDD induction, this has been demonstrated to promote disruption and persistent changes to chromatin structure [9, 10], enhance cellular senescence [11], and to increase apoptotic signaling [12]. This reflects, to some degree, the potentially diverse mechanisms through which high-LET radiation may act

Objectives
Methods
Results
Conclusion
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