Quantification of ongoing APOBEC3A activity in tumor cells by monitoring RNA editing at hotspots

Autor: Michael S. Lawrence, Angela G. Fleischman, Kevin Aguirre, Shinho Park, Adam Langenbucher, Ryan B. Corcoran, Lee Zou, Danae Bowen, Pégah Jalili, Rémi Buisson
Jazyk: angličtina
Rok vydání: 2020
Předmět:
0301 basic medicine
General Physics and Astronomy
Biochemistry
Whole Exome Sequencing
0302 clinical medicine
RNA interference
Neoplasms
2.1 Biological and endogenous factors
Digital polymerase chain reaction
Aetiology
APOBEC3A
lcsh:Science
Cancer
Multidisciplinary
Tumor
Biological techniques
Cytidine deaminase
Gene Expression Regulation
Neoplastic

RNA editing
030220 oncology & carcinogenesis
RNA Interference
Biotechnology
DNA damage
Science
Biology
General Biochemistry
Genetics and Molecular Biology

Article
Cell Line
03 medical and health sciences
Cell Line
Tumor

Cytidine Deaminase
Exome Sequencing
Genetics
Humans
Enzyme Assays
Neoplastic
RNA
Proteins
General Chemistry
Computational biology and bioinformatics
030104 developmental biology
HEK293 Cells
Gene Expression Regulation
Cancer cell
Mutation
Cancer research
lcsh:Q
RNA Editing
Zdroj: Nature Communications, Vol 11, Iss 1, Pp 1-13 (2020)
Nature Communications
Nature communications, vol 11, iss 1
ISSN: 2041-1723
Popis: APOBEC3A is a cytidine deaminase driving mutagenesis, DNA replication stress and DNA damage in cancer cells. While the APOBEC3A-induced vulnerability of cancers offers an opportunity for therapy, APOBEC3A protein and mRNA are difficult to quantify in tumors due to their low abundance. Here, we describe a quantitative and sensitive assay to measure the ongoing activity of APOBEC3A in tumors. Using hotspot RNA mutations identified from APOBEC3A-positive tumors and droplet digital PCR, we develop an assay to quantify the RNA-editing activity of APOBEC3A. This assay is superior to APOBEC3A protein- and mRNA-based assays in predicting the activity of APOBEC3A on DNA. Importantly, we demonstrate that the RNA mutation-based APOBEC3A assay is applicable to clinical samples from cancer patients. Our study presents a strategy to follow the dysregulation of APOBEC3A in tumors, providing opportunities to investigate the role of APOBEC3A in tumor evolution and to target the APOBEC3A-induced vulnerability in therapy.
The DNA cytosine deaminases APOBEC3A and APOBEC3B have emerged from cancer genomics studies as drivers of mutation in cancers and tumor heterogeneity. Here the authors present a computational approach to identify the RNA mutations specifically driven by APOBEC3A, and developed an RNA mutation-based assay to quantify ongoing APOBEC3A activity in tumor cells.
Databáze: OpenAIRE