4.7 Article

DEK is required for homologous recombination repair of DNA breaks

Journal

SCIENTIFIC REPORTS
Volume 7, Issue -, Pages -

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/srep44662

Keywords

-

Funding

  1. three NIH P30 grants through the Division of Rheumatology's Center for Rheumatic Disease Research [NIH AR47363]
  2. Division of Gastroenterology's Digestive Health Center [NIH DK78392]
  3. Division of Experimental Hematology and Cancer Biology's Center for Excellence in Molecular Hematology [NIH DK90971]
  4. National Institutes of Health [R01-CA-102357, R01-CA116316, R01-CA095175, T32 GM063483-13, T32-ES007250]
  5. German Research Foundation (DFG) [WI 3099/7-1, WI 3099/7-2, KA 2799/1]
  6. RWTH Aachen START Program of the Faculty of Medicine
  7. University of Cincinnati Department of Cancer and Cell Biology Cardell Fellowship for Excellence in Graduate Research
  8. Chinese Scholarship Council
  9. German Cancer Aid (Deutsche Krebshilfe) [110388]
  10. Novo Nordisk Foundation [NNF14CC0001]
  11. Danish Council for Independent Research in Medical Sciences

Ask authors/readers for more resources

DEK is a highly conserved chromatin-bound protein whose upregulation across cancer types correlates with genotoxic therapy resistance. Loss of DEK induces genome instability and sensitizes cells to DNA double strand breaks (DSBs), suggesting defects in DNA repair. While these DEK-deficiency phenotypes were thought to arise from a moderate attenuation of non-homologous end joining (NHEJ) repair, the role of DEK in DNA repair remains incompletely understood. We present new evidence demonstrating the observed decrease in NHEJ is insufficient to impact immunoglobulin class switching in DEK knockout mice. Furthermore, DEK knockout cells were sensitive to apoptosis with NHEJ inhibition. Thus, we hypothesized DEK plays additional roles in homologous recombination (HR). Using episomal and integrated reporters, we demonstrate that HR repair of conventional DSBs is severely compromised in DEK-deficient cells. To define responsible mechanisms, we tested the role of DEK in the HR repair cascade. DEK-deficient cells were impaired for gamma H2AX phosphorylation and attenuated for RAD51 filament formation. Additionally, DEK formed a complex with RAD51, but not BRCA1, suggesting a potential role regarding RAD51 filament formation, stability, or function. These findings define DEK as an important and multifunctional mediator of HR, and establish a synthetic lethal relationship between DEK loss and NHEJ inhibition.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available