Identification of Aging-Associated Gene Expression Signatures That Precede Intestinal Tumorigenesis

PLoS One. 2016 Sep 2;11(9):e0162300. doi: 10.1371/journal.pone.0162300. eCollection 2016.

Abstract

Aging-associated alterations of cellular functions have been implicated in various disorders including cancers. Due to difficulties in identifying aging cells in living tissues, most studies have focused on aging-associated changes in whole tissues or certain cell pools. Thus, it remains unclear what kinds of alterations accumulate in each cell during aging. While analyzing several mouse lines expressing fluorescent proteins (FPs), we found that expression of FPs is gradually silenced in the intestinal epithelium during aging in units of single crypt composed of clonal stem cell progeny. The cells with low FP expression retained the wild-type Apc allele and the tissues composed of them did not exhibit any histological abnormality. Notably, the silencing of FPs was also observed in intestinal adenomas and the surrounding normal mucosae of Apc-mutant mice, and mediated by DNA methylation of the upstream promoter. Our genome-wide analysis then showed that the silencing of FPs reflects specific gene expression alterations during aging, and that these alterations occur in not only mouse adenomas but also human sporadic and hereditary (familial adenomatous polyposis) adenomas. Importantly, pharmacological inhibition of DNA methylation, which suppresses adenoma development in Apc-mutant mice, reverted the aging-associated silencing of FPs and gene expression alterations. These results identify aging-associated gene expression signatures that are heterogeneously induced by DNA methylation and precede intestinal tumorigenesis triggered by Apc inactivation, and suggest that pharmacological inhibition of the signature genes could be a novel strategy for the prevention and treatment of intestinal tumors.

MeSH terms

  • Adenoma / genetics*
  • Adenoma / metabolism
  • Adenoma / pathology
  • Aging / genetics*
  • Aging / metabolism
  • Animals
  • Carcinogenesis / genetics*
  • Cell Transformation, Neoplastic / genetics*
  • Cell Transformation, Neoplastic / metabolism
  • Cell Transformation, Neoplastic / pathology
  • Gene Expression Regulation, Neoplastic
  • Gene Expression*
  • Genetic Markers
  • Humans
  • Intestinal Mucosa / metabolism
  • Intestinal Neoplasms / genetics*
  • Intestinal Neoplasms / metabolism
  • Mice

Substances

  • Genetic Markers

Grants and funding

YO and MM were supported by the Platform Project for Supporting Drug Discovery and Life Science Research (Platform for Dynamic Approaches to Living System) from the Ministry of Education, Culture, Sports, and Science (MEXT) and the Japan Agency for Medical Research and Development (AMED) (http://www.jst.go.jp/EN/index.html), and by a Grant-in-Aid for Scientific Research on Innovative Areas "Resonance Biology" of MEXT (https://kaken.nii.ac.jp/grant/KAKENHI-PLANNED-15H05949/). MM was also supported by the Naito Foundation (https://www.naito-f.or.jp/en/). MI was supported by a Grant-in-Aid for Young Scientists (B) (25870363) (https://kaken.nii.ac.jp/en/grant/KAKENHI-PROJECT-25870363/). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.