Genome Biology (Jun 2021)

Highly recurrent CBS epimutations in gastric cancer CpG island methylator phenotypes and inflammation

  • Nisha Padmanabhan,
  • Huang Kie Kyon,
  • Arnoud Boot,
  • Kevin Lim,
  • Supriya Srivastava,
  • Shuwen Chen,
  • Zhiyuan Wu,
  • Hyung-O K Lee,
  • Vineeth T. Mukundan,
  • Charlene Chan,
  • Yarn Kit Chan,
  • Ong Xuewen,
  • Jason J. Pitt,
  • Zul Fazreen Adam Isa,
  • Manjie Xing,
  • Ming Hui Lee,
  • Angie Lay Keng Tan,
  • Shamaine Ho Wei Ting,
  • Micah A. Luftig,
  • Dennis Kappei,
  • Warren D. Kruger,
  • Jinsong Bian,
  • Ying Swan Ho,
  • Ming Teh,
  • Steve George Rozen,
  • Patrick Tan

DOI
https://doi.org/10.1186/s13059-021-02375-2
Journal volume & issue
Vol. 22, no. 1
pp. 1 – 29

Abstract

Read online

Abstract Background CIMP (CpG island methylator phenotype) is an epigenetic molecular subtype, observed in multiple malignancies and associated with the epigenetic silencing of tumor suppressors. Currently, for most cancers including gastric cancer (GC), mechanisms underlying CIMP remain poorly understood. We sought to discover molecular contributors to CIMP in GC, by performing global DNA methylation, gene expression, and proteomics profiling across 14 gastric cell lines, followed by similar integrative analysis in 50 GC cell lines and 467 primary GCs. Results We identify the cystathionine beta-synthase enzyme (CBS) as a highly recurrent target of epigenetic silencing in CIMP GC. Likewise, we show that CBS epimutations are significantly associated with CIMP in various other cancers, occurring even in premalignant gastroesophageal conditions and longitudinally linked to clinical persistence. Of note, CRISPR deletion of CBS in normal gastric epithelial cells induces widespread DNA methylation changes that overlap with primary GC CIMP patterns. Reflecting its metabolic role as a gatekeeper interlinking the methionine and homocysteine cycles, CBS loss in vitro also causes reductions in the anti-inflammatory gasotransmitter hydrogen sulfide (H2S), with concomitant increase in NF-κB activity. In a murine genetic model of CBS deficiency, preliminary data indicate upregulated immune-mediated transcriptional signatures in the stomach. Conclusions Our results implicate CBS as a bi-faceted modifier of aberrant DNA methylation and inflammation in GC and highlights H2S donors as a potential new therapy for CBS-silenced lesions.

Keywords