The deaminase APOBEC3B triggers the death of cells lacking uracil DNA glycosylase

Human cells express up to 9 active DNA cytosine deaminases with functions in adaptive and innate immunity. Many cancers manifest an APOBEC mutation signature and APOBEC3B (A3B) is likely the main enzyme responsible. Although significant numbers of APOBEC signature mutations accumulate in tumor genom...

Full description

Saved in:
Bibliographic Details
Published inProceedings of the National Academy of Sciences - PNAS Vol. 116; no. 44; pp. 22158 - 22163
Main Authors Serebrenik, Artur A., Starrett, Gabriel J., Leenen, Sterre, Jarvis, Matthew C., Shaban, Nadine M., Salamango, Daniel J., Nilsen, Hilde, Brown, William L., Harris, Reuben S.
Format Journal Article
LanguageEnglish
Published United States National Academy of Sciences 29.10.2019
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Human cells express up to 9 active DNA cytosine deaminases with functions in adaptive and innate immunity. Many cancers manifest an APOBEC mutation signature and APOBEC3B (A3B) is likely the main enzyme responsible. Although significant numbers of APOBEC signature mutations accumulate in tumor genomes, the majority of APOBEC-catalyzed uracil lesions are probably counteracted in an error-free manner by the uracil base excision repair pathway. Here, we show that A3B-expressing cells can be selectively killed by inhibiting uracil DNA glycosylase 2 (UNG) and that this synthetic lethal phenotype requires functional mismatch repair (MMR) proteins and p53. UNG knockout human 293 and MCF10A cells elicit an A3B-dependent death. This synthetic lethal phenotype is dependent on A3B catalytic activity and reversible by UNG complementation. A3B expression in UNG-null cells causes a buildup of genomic uracil, and the ensuing lethality requires processing of uracil lesions (likely U/G mispairs) by MSH2 and MLH1 (likely noncanonical MMR). Cancer cells expressing high levels of endogenous A3B and functional p53 can also be killed by expressing an UNG inhibitor. Taken together, UNG-initiated base excision repair is a major mechanism counteracting genomic mutagenesis by A3B, and blocking UNG is a potential strategy for inducing the selective death of tumors.
Bibliography:Edited by Richard D. Kolodner, Ludwig Institute for Cancer Research, La Jolla, CA, and approved September 24, 2019 (received for review March 21, 2019)
Author contributions: A.A.S., H.N., and R.S.H. designed research; A.A.S., G.J.S., S.L., M.C.J., N.M.S., D.J.S., and W.L.B. performed research; W.L.B. contributed new reagents/analytic tools; A.A.S., G.J.S., S.L., M.C.J., N.M.S., D.J.S., H.N., and R.S.H. analyzed data; and A.A.S. and R.S.H. wrote the paper.
ISSN:0027-8424
1091-6490
DOI:10.1073/pnas.1904024116