Your browser doesn't support javascript.
loading
DNA methyltransferase inhibition reduces inflammation-induced colon tumorigenesis.
Maiuri, Ashley R; Savant, Sudha S; Podicheti, Ram; Rusch, Douglas B; O'Hagan, Heather M.
Affiliation
  • Maiuri AR; Medical Sciences, Indiana University School of Medicine , Bloomington , IN , USA.
  • Savant SS; Medical Sciences, Indiana University School of Medicine , Bloomington , IN , USA.
  • Podicheti R; School of Informatics, Computing and Engineering, Indiana University , Bloomington , IN , USA.
  • Rusch DB; Center for Genomics and Bioinformatics, Indiana University , Bloomington , IN , USA.
  • O'Hagan HM; Center for Genomics and Bioinformatics, Indiana University , Bloomington , IN , USA.
Epigenetics ; 14(12): 1209-1223, 2019 12.
Article in En | MEDLINE | ID: mdl-31240997
ABSTRACT
Chronic inflammation is strongly associated with an increased risk of developing colorectal cancer. DNA hypermethylation of CpG islands alters the expression of genes in cancer cells and plays an important role in carcinogenesis. Chronic inflammation is also associated with DNA methylation alterations and in a mouse model of inflammation-induced colon tumorigenesis, we previously demonstrated that inflammation-induced tumours have 203 unique regions with DNA hypermethylation compared to uninflamed epithelium. To determine if altering inflammation-induced DNA hypermethylation reduces tumorigenesis, we used the same mouse model and treated mice with the DNA methyltransferase (DNMT) inhibitor decitabine (DAC) throughout the tumorigenesis time frame. DAC treatment caused a significant reduction in colon tumorigenesis. The tumours that did form after DAC treatment had reduced inflammation-specific DNA hypermethylation and alteration of expression of associated candidate genes. When compared, inflammation-induced tumours from control (PBS-treated) mice were enriched for cell proliferation associated gene expression pathways whereas inflammation-induced tumours from DAC-treated mice were enriched for interferon gene signatures. To further understand the altered tumorigenesis, we derived tumoroids from the different tumour types. Interestingly, tumoroids derived from inflammation-induced tumours from control mice maintained many of the inflammation-induced DNA hypermethylation alterations and had higher levels of DNA hypermethylation at these regions than tumoroids from DAC-treated mice. Importantly, tumoroids derived from inflammation-induced tumours from the DAC-treated mice proliferated more slowly than those derived from the inflammation-induced tumours from control mice. These studies suggest that inhibition of inflammation-induced DNA hypermethylation may be an effective strategy to reduce inflammation-induced tumorigenesis.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: DNA-Cytosine Methylases / Colonic Neoplasms / DNA Methylation / Carcinogenesis Limits: Animals Language: En Journal: Epigenetics Journal subject: GENETICA Year: 2019 Document type: Article Affiliation country: Estados Unidos

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: DNA-Cytosine Methylases / Colonic Neoplasms / DNA Methylation / Carcinogenesis Limits: Animals Language: En Journal: Epigenetics Journal subject: GENETICA Year: 2019 Document type: Article Affiliation country: Estados Unidos