دورية أكاديمية

Elevated APOBEC3B expression drives a kataegic-like mutation signature and replication stress-related therapeutic vulnerabilities in p53-defective cells.

التفاصيل البيبلوغرافية
العنوان: Elevated APOBEC3B expression drives a kataegic-like mutation signature and replication stress-related therapeutic vulnerabilities in p53-defective cells.
المؤلفون: Nikkilä, J, Kumar, R, Campbell, J, Brandsma, I, Pemberton, HN, Wallberg, F, Nagy, K, Scheer, I, Vertessy, BG, Serebrenik, AA, Monni, V, Harris, RS, Pettitt, SJ, Ashworth, A, Lord, CJ
المساهمون: Campbell, James, Pettitt, Stephen, Lord, Christopher
بيانات النشر: NATURE PUBLISHING GROUP
سنة النشر: 2017
المجموعة: The Institute of Cancer Research (ICR): Publications Repository
مصطلحات موضوعية: Cell Line, Humans, DNA Damage, Cisplatin, Uracil, Cytidine Deaminase, Cell Cycle Proteins, Nuclear Proteins, Minor Histocompatibility Antigens, Blotting, Western, Gene Expression Regulation, Neoplastic, RNA Interference, Mutation, Tumor Suppressor Protein p53, Protein-Tyrosine Kinases, Gene Knockout Techniques, HEK293 Cells, Cell Cycle Checkpoints, G2 Phase Cell Cycle Checkpoints, Ataxia Telangiectasia Mutated Proteins, Checkpoint Kinase 2, CRISPR-Cas Systems, Poly(ADP-ribose) Polymerase Inhibitors, Checkpoint Kinase 1
الوصف: BACKGROUND: Elevated APOBEC3B expression in tumours correlates with a kataegic pattern of localised hypermutation. We assessed the cellular phenotypes associated with high-level APOBEC3B expression and the influence of p53 status on these phenotypes using an isogenic system. METHODS: We used RNA interference of p53 in cells with inducible APOBEC3B and assessed DNA damage response (DDR) biomarkers. The mutational effects of APOBEC3B were assessed using whole-genome sequencing. In vitro small-molecule inhibitor sensitivity profiling was used to identify candidate therapeutic vulnerabilities. RESULTS: Although APOBEC3B expression increased the incorporation of genomic uracil, invoked DDR biomarkers and caused cell cycle arrest, inactivation of p53 circumvented APOBEC3B-induced cell cycle arrest without reversing the increase in genomic uracil or DDR biomarkers. The continued expression of APOBEC3B in p53-defective cells not only caused a kataegic mutational signature but also caused hypersensitivity to small-molecule DDR inhibitors (ATR, CHEK1, CHEK2, PARP, WEE1 inhibitors) as well as cisplatin/ATR inhibitor and ATR/PARP inhibitor combinations. CONCLUSIONS: Although loss of p53 might allow tumour cells to tolerate elevated APOBEC3B expression, continued expression of this enzyme might impart a number of therapeutic vulnerabilities upon tumour cells.
نوع الوثيقة: article in journal/newspaper
وصف الملف: Print-Electronic; 123; application/pdf
اللغة: English
تدمد: 0007-0920
1532-1827
العلاقة: British journal of cancer, 2017, 117 (1), pp. 113 - 123; https://repository.icr.ac.uk/handle/internal/850Test
DOI: 10.1038/bjc.2017.133
الإتاحة: https://doi.org/10.1038/bjc.2017.133Test
https://repository.icr.ac.uk/handle/internal/850Test
حقوق: https://creativecommons.org/licenses/by/4.0Test
رقم الانضمام: edsbas.322BE020
قاعدة البيانات: BASE
الوصف
تدمد:00070920
15321827
DOI:10.1038/bjc.2017.133