Gastroenterology

Gastroenterology

Volume 151, Issue 4, October 2016, Pages 637-650.e10
Gastroenterology

Original Research
Full Report: Clinical—Alimentary Tract
ADAR-Mediated RNA Editing Predicts Progression and Prognosis of Gastric Cancer

https://doi.org/10.1053/j.gastro.2016.06.043Get rights and content

Backgroud & Aims

Gastric cancer (GC) is the third leading cause of global cancer mortality. Adenosine-to-inosine RNA editing is a recently described novel epigenetic mechanism involving sequence alterations at the RNA but not DNA level, primarily mediated by ADAR (adenosine deaminase that act on RNA) enzymes. Emerging evidence suggests a role for RNA editing and ADARs in cancer, however, the relationship between RNA editing and GC development and progression remains unknown.

Methods

In this study, we leveraged on the next-generation sequencing transcriptomics to demarcate the GC RNA editing landscape and the role of ADARs in this deadly malignancy.

Results

Relative to normal gastric tissues, almost all GCs displayed a clear RNA misediting phenotype with ADAR1/2 dysregulation arising from the genomic gain and loss of the ADAR1 and ADAR2 gene in primary GCs, respectively. Clinically, patients with GCs exhibiting ADAR1/2 imbalance demonstrated extremely poor prognoses in multiple independent cohorts. Functionally, we demonstrate in vitro and in vivo that ADAR-mediated RNA misediting is closely associated with GC pathogenesis, with ADAR1 and ADAR2 playing reciprocal oncogenic and tumor suppressive roles through their catalytic deaminase domains, respectively. Using an exemplary target gene PODXL (podocalyxin-like), we demonstrate that the ADAR2-regulated recoding editing at codon 241 (His to Arg) confers a loss-of-function phenotype that neutralizes the tumorigenic ability of the unedited PODXL.

Conclusions

Our study highlights a major role for RNA editing in GC disease and progression, an observation potentially missed by previous next-generation sequencing analyses of GC focused on DNA alterations alone. Our findings also suggest new GC therapeutic opportunities through ADAR1 enzymatic inhibition or the potential restoration of ADAR2 activity.

Section snippets

Materials and Methods

The detailed Materials and Methods can be found in the Supplementary Materials.

Global Identification of Adenosine-to-Inosine/Guanosine (A-to-I or A-to-G) RNA Editing Sites in Gastric Cancer by RNA-Seq

We performed high-throughput RNA-Seq of 14 matched pairs of gastric tumors and non-tumor (NT) gastric samples, generating a mean of 170.3-million reads that could be uniquely aligned to the human genome (hg19). The aligned reads provided substantial coverage (a mean of 68.1%) for the vast majority of human messenger RNA (mRNA) transcripts annotated in the UCSC genome browser (Supplementary Table 1). In order to perform a comprehensive high-quality analysis, we applied a 2-phase method for

Discussion

This study reports a comprehensive transcriptome-wide analysis of the GC RNA editing landscape, and a clinically significant relationship between ADAR-mediated RNA editing and GC pathogenesis and progression. Previous molecular studies of GC have focused primarily on characterizing aberrations at the DNA level, such as mutations, copy number amplifications, and fusion genes.3, 4, 5, 6, 7, 8, 9, 10 Alternatively, while GC gene expression studies using microarrays and RNA sequencing have been

Acknowledgments

The authors thank Won Ki Kang, Sung Kim, and Hyun Cheol Cheong for contributing to clinical data and reagents associated with this project. We acknowledge the contributions of the Duke-NUS Genome Biology Facility for genomic profiling services.

Accession code: The RNA-Seq data were deposited in the following repository: Repository/DataBank Accession: European Genome-phenome Archive, accession no: EGAS00001001128. Databank URL: https://ega.crg.eu/.

References (54)

  • L. Han et al.

    The genomic landscape and clinical relevance of A-to-I RNA editing in human cancers

    Cancer Cell

    (2015)
  • N. Paz-Yaacov et al.

    Elevated RNA editing activity is a major contributor to transcriptomic diversity in tumors

    Cell Rep

    (2015)
  • D. Fumagalli et al.

    Principles governing A-to-I RNA editing in the breast cancer transcriptome

    Cell Rep

    (2015)
  • D. Carroll

    A CRISPR approach to gene targeting

    Mol Ther

    (2012)
  • S.S. Wohrer et al.

    Palliative chemotherapy for advanced gastric cancer

    Ann Oncol

    (2004)
  • D.M. Parkin et al.

    Global cancer statistics, 2002

    CA Cancer J Clin

    (2005)
  • K. Wang et al.

    Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer

    Nat Genet

    (2014)
  • J.G. Strickler et al.

    p53 mutations and microsatellite instability in sporadic gastric cancer: when guardians fail

    Cancer Res

    (1994)
  • M.H. McLean et al.

    Genetics of gastric cancer

    Nat Rev Gastroenterol Hepatol

    (2014)
  • J. Lee et al.

    Identification of ROS1 rearrangement in gastric adenocarcinoma

    Cancer

    (2013)
  • J. Tao et al.

    CD44-SLC1A2 gene fusions in gastric cancer

    Sci Transl Med

    (2011)
  • N. Boku

    HER2-positive gastric cancer

    Gastric Cancer

    (2014)
  • H. Zouridis et al.

    Methylation subtypes and large-scale epigenetic alterations in gastric cancer

    Sci Transl Med

    (2012)
  • S.Y. Park et al.

    CpG island hypermethylator phenotype in gastric carcinoma and its clinicopathological features

    Virchows Arch

    (2010)
  • L. Qi et al.

    RNA editome imbalance in hepatocellular carcinoma

    Cancer Res

    (2014)
  • A. Athanasiadis et al.

    Widespread A-to-I RNA editing of Alu-containing mRNAs in the human transcriptome

    PLoS Biol

    (2004)
  • K. Nishikura

    Editor meets silencer: crosstalk between RNA editing and RNA interference

    Nat Rev Mol Cell Biol

    (2006)
  • Cited by (0)

    Conflicts of interest The authors disclose no conflicts.

    Funding This work was supported by the National Research Foundation Singapore, and the Singapore Ministry of Education under its Research Centres of Excellence initiative, NMRC Clinician Scientist-Individual Research Grant New Investigator Grant (CS-IRG NIG, grant number: NMRC/CNIG/1117/2014); NMRC Clinician Scientist-Individual Research Grant (CS-IRG, grant number: NMRC/CIRG/1412/2014); NUS Young Investigator Award (NUS YIA, grant number: NUSYIA_FY14_P22), NUS Start-up Fund (Ref number: NUHSRO/2015/095/SU/01); and core grants from Duke-NUS, GIS, and NMRC/TCR/009-NUHS/2013. The latter was supported through the National Medical Research Council/National Research Foundation Translational and Clinical Research (TCR) Flagship Program. This research is also supported by the RNA Biology Center at the Cancer Science Institute of Singapore, NUS, as part of funding under the Singapore Ministry of Education’s Tier 3 grants (MOE2014-T3-1-006).

    Author names in bold designate shared co-first authorship.

    View full text