Epigenetic silencing of erythropoietin in human cancers

K Steinmann, AM Richter, RH Dammann - Genes & cancer, 2011 - journals.sagepub.com
K Steinmann, AM Richter, RH Dammann
Genes & cancer, 2011journals.sagepub.com
The glycoprotein hormone erythropoietin (EPO) is a key regulator in the production of red
blood cells. EPO is produced mainly in the embryonic liver and kidney of adults. Other
organs are also known to express varying amounts of EPO. In our study, we have analyzed
the epigenetic regulation of EPO in human cancer cell lines by DNA methylation assays,
chromatin immunoprecipitation, RT-PCR, and promoter analysis under different growth
conditions. Moreover, the growth-related effects of ectopic EPO expression were analyzed in …
The glycoprotein hormone erythropoietin (EPO) is a key regulator in the production of red blood cells. EPO is produced mainly in the embryonic liver and kidney of adults. Other organs are also known to express varying amounts of EPO. In our study, we have analyzed the epigenetic regulation of EPO in human cancer cell lines by DNA methylation assays, chromatin immunoprecipitation, RT-PCR, and promoter analysis under different growth conditions. Moreover, the growth-related effects of ectopic EPO expression were analyzed in a head and neck cancer cell line. We found frequent DNA hypermethylation of the CpG island promoter and enhancer of EPO in different cancer cell lines. Aberrant methylation of EPO promoter was observed in primary lung, head and neck, breast, and liver cancers. Hypermethylation of EPO was associated with a decreased expression of EPO in cancer cells. Treatment of cancer cell lines with 5-aza-2′-deoxycytidine (Aza), an inhibitor of DNA methylation, reactivated EPO expression under hypoxia. In contrast, in the liver cancer cell line HepB3, the EPO promoter was unmethylated, and a high EPO expression was observed independently of Aza treatment. Moreover, in vitro hypermethylation of the EPO promoter and enhancer reduced expression of a reporter gene under normoxia and hypoxia. Induction of EPO under hypoxia was accompanied by increased histone H3 acetylation and reduced histone H3 lysine 9 trimethylation. In a head and neck cancer cell line, which exhibited low EPO levels, ectopic expression of EPO significantly enhanced proliferation under normoxia and hypoxia. In summary, we show that hypermethylation of regulatory sequences of EPO is frequently observed in tumors and that this aberrant methylation induces epigenetic silencing of EPO in cancer cells.
Sage Journals