Skip Navigation

Publication Detail

Title: Upregulation of histone-lysine methyltransferases plays a causal role in hexavalent chromium-induced cancer stem cell-like property and cell transformation.

Authors: Wang, Zhishan; Wu, Jianjun; Humphries, Brock; Kondo, Kazuya; Jiang, Yiguo; Shi, Xianglin; Yang, Chengfeng

Published In Toxicol Appl Pharmacol, (2018 Mar 01)

Abstract: While hexavalent chromium [Cr(VI)] is generally considered as a genotoxic environmental carcinogen, studies showed that Cr(VI) exposure also causes epigenetic changes. However, whether Cr(VI)-caused epigenetic dysregulations plays an important role in Cr(VI) carcinogenicity remain largely unknown. The aim of this study was to determine if chronic low dose Cr(VI) exposure causes epigenetic changes, the underlying mechanism and whether chronic low dose Cr(VI) exposure-caused epigenetic dysregulation contributes causally to Cr(VI)-induced cancer stem cell (CSC)-like property and cell transformation. Two immortalized human bronchial epithelial cell lines (BEAS-2B and 16HBE) were exposed to 0.25 μM of K2Cr2O7 for 20 and 40 weeks to induce cell transformation, respectively. Cr(VI)-induced epigenetic changes were examined in Cr(VI)-transformed cells and Cr(VI) exposure-caused human lung cancer tissues. Pharmacological inhibitors and gene knockdown experiments were used to determine the role of epigenetic dysregulation in Cr(VI) carcinogenicity. We found that chronic Cr(VI) exposure causes epigenetic dysregulation as evidenced by the increased levels of histone H3 repressive methylation marks (H3K9me2 and H3K27me3) and the related histone-lysing methyltransferases (HMTases). Pharmacological inhibition or knockdown of HMTases reduces H3 repressive methylation marks and malignant phenotypes of Cr(VI)-transformed cells. Moreover, knockdown of HMTases in parental cells significantly reduces chronic Cr(VI) exposure-induced CSC-like property and cell transformation. Further mechanistic study revealed that knockdown of HMTases decreases Cr(VI) exposure-caused DNA damage. Our findings indicate that chronic Cr(VI) exposure increases H3 repressive methylation marks by increasing the related HMTases expression; and that increased expression of HMTases plays a causal role in Cr(VI)-induced CSC-like property and cell transformation.

PubMed ID: 29391238 Exiting the NIEHS site

MeSH Terms: Animals; Cell Line, Transformed; Cell Transformation, Neoplastic/drug effects; Cell Transformation, Neoplastic/metabolism*; Cell Transformation, Neoplastic/pathology; Chromium/toxicity*; Histone-Lysine N-Methyltransferase/biosynthesis*; Humans; Mice; Mice, Nude; Neoplastic Stem Cells/drug effects; Neoplastic Stem Cells/metabolism*; Neoplastic Stem Cells/pathology; Up-Regulation/drug effects; Up-Regulation/physiology*; Xenograft Model Antitumor Assays/methods

Back
to Top