Published in

American Association for Cancer Research, Cancer Research, 13_Supplement(78), p. 2993-2993, 2018

DOI: 10.1158/1538-7445.am2018-2993

Links

Tools

Export citation

Search in Google Scholar

Abstract 2993: A switch in epigenetic silencing mechanisms of endogenous retroviruses during human genome evolution

Journal article published in 2018 by Hitoshi Ohtani, Minmin Liu, Wanding Zhou, Gangning Liang, Peter A. Jones
This paper was not found in any repository, but could be made available legally by the author.
This paper was not found in any repository, but could be made available legally by the author.

Full text: Unavailable

Green circle
Preprint: archiving allowed
Orange circle
Postprint: archiving restricted
Red circle
Published version: archiving forbidden
Data provided by SHERPA/RoMEO

Abstract

Abstract Approximately 9% of the human genome is derived from Endogenous Retroviruses (ERVs), which are a type of transposable element. ERVs started to become integrated into the ancestral human genome about 100 million years ago. Recent studies have reported that silencing mechanisms of long terminal repeats regions (LTRs), which are the putative promoters of ERVs, depend on DNA methylation, heterochromatin conformation (H3K9me3) and the PRC2 complex (H3K27me3). However, extensive maps of LTR distribution and the associated epigenetic marks have not yet been provided in human cancer cells. Here we report a switch in epigenetic silencing mechanisms of the approximately 450,000 LTR elements present in the non-genic regions of human DNA. Total RNA-seq in human colon cancer HCT116 cells following DNA methylation inhibitor treatment or knockdown of individual H3K9me2/3 histone methyltransferases (G9a, SETDB1, SETDB2, SUV39H1 and SUV39H2), H3K27me3 (EZH2) or a scaffold protein TRIM28 revealed that about 1,000 of evolutionary young LTRs were predominantly silenced by DNA methylation, whereas about 800 of intermediate age LTRs were silenced by histone methylation. They therefore have undergone an “epigenetic switch” in silencing mechanism during host genome evolution. Eventually, evolutionary old LTRs become inactive by accumulation of loss-of-function mutations. Consistent with these results, CpG sites in evolutionary young LTRs were methylated at higher levels than in intermediate age LTRs, whereas H3K9me2 and H3K9me3 marks were enriched on intermediate age LTRs. Interestingly, knockdowns of EZH2 showed limited upregulation of LTRs by itself, but multiple LTRs were upregulated by combination treatment with DNA methylation inhibitor. This is probably due to a process, in which removal of DNA methylation leads to the subsequent recruitment of EZH2 resulting in suppression by the polycomb repressive complex PRC2. Our earlier studies showed that LTR expressions lead to activation of viral defense pathways and apoptosis, however the species of LTRs was not characterized. We found that the activation of viral defense pathway was correlated with the upregulation of evolutionary young, but not intermediate age LTRs. Dual inhibition of DNA methylation and histone modifications by knockdown of G9a, SUV39H1 and EZH2 showed further upregulation of evolutionary young LTRs. Therefore, combination treatment targeting DNA methylation and the histone methyltransferases (G9a, SUV39H1 and EZH2) inhibitor may be useful in epigenetic therapy for cancer patients. Citation Format: Hitoshi Ohtani, Minmin Liu, Wanding Zhou, Gangning Liang, Peter A. Jones. A switch in epigenetic silencing mechanisms of endogenous retroviruses during human genome evolution [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2993.