4.5 Article

Identification of Diverse Adenosine-to-Inosine RNA Editing Subtypes in Colorectal Cancer

Journal

CANCER RESEARCH AND TREATMENT
Volume 49, Issue 4, Pages 1077-1087

Publisher

KOREAN CANCER ASSOCIATION
DOI: 10.4143/crt.2016.301

Keywords

RNA editing; Colorectal neoplasms; Transcriptome sequencing; GLI family zinc finger 1; Adenosine deaminase

Categories

Funding

  1. Korea Health Technology R&D Project through the Korea Health Industry Development Institute - Ministry of Health Welfare [HI14C1277]
  2. Priority Research Centers Program through the NRF - Ministry of Education, Science and Technology, Republic of Korea [2009-0093820]
  3. Doosan Yongkang Foundation [30-2013-015-0]
  4. Ministry of Science, ICT & Future Planning [2016M3A9B6026918]
  5. National Research Foundation of Korea [2016M3A9B6026918] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)

Ask authors/readers for more resources

Purpose RNA editing generates protein diversity by altering RNA sequences in coding regions without changing the overall DNA sequence. Adenosine-to-inosine (A-to-I) RNA editing events have recently been reported in some types of cancer, but they are rare in human colorectal cancer (CRC). Therefore, this study was conducted to identify diverse RNA editing in CRC. Materials and Methods We compared transcriptome data of 39 CRC samples and paired adjacent tissues from The Cancer Genome Atlas database to identify RNA editing patterns in CRC, focusing on canonical A-to-I RNA edits in coding sequence regions. We investigated nonsynonymous RNA editing patterns by comparing tumorand normal tissue transcriptome data. Results The number of RNA edits varied from 12 to 42 per sample. We also observed that hypo-and hyper-RNA editing patterns were distinguishable within the samples. We found 10 recurrent nonsynonymous RNA editing candidates in nine genes (PDLIM, NEIL1, SRP9, GLI1, APMAP, IGFBP7, ZNF358, COPA, and ZNF587B) and validated some by Sanger sequencing and the inosine chemical erasing assay. We further showed that editing at these positions was performed by the adenosine deaminase acting on RNA 1 enzyme. Most of these genes are hypoedited in CRC, but editing of GLI1 was increased in cancer tissues compared with normal tissues. Conclusion Our results show that nonsynonymous RNA editing patterns can be used to identify CRC patients and could serve as novel biomarkers for CRC.

Authors

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

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available