Inhibition of the HDAC/Suv39/G9a pathway restores the expression of DNA damage-dependent major histocompatibility complex class I-related chain A and B in cancer cells

Oncol Rep. 2017 Aug;38(2):693-702. doi: 10.3892/or.2017.5773. Epub 2017 Jun 30.

Abstract

Immunotherapy is expected to be promising as a next generation cancer therapy. Immunoreceptors are often activated constitutively in cancer cells, however, such levels of ligand expression are not effectively recognized by the native immune system due to tumor microenvironmental adaptation. Studies have demonstrated that natural-killer group 2, member D (NKG2D), a major activating immunoreceptor, responds to DNA damage. The upregulation of major histocompatibility complex class I-related chain A and B (MICA/B) (members of NKG2D ligands) expression after DNA damage is associated with NK cell-mediated killing of cancer cells. However, the regulation of DNA damage-induced MICA/B expression has not been fully elucidated in the context of the types of cancer cell lines. In the present study, we found that MICA/B expression varied between cancer cell lines after DNA damage. Screening in terms of chromatin remodeling identified that inhibitors related to chromatin relaxation via post-translational modification on histone H3K9, i.e. HDAC, Suv39 or G9a inhibition, restored DNA damage-dependent MICA/B expression in insensitive cells. In addition, we revealed that the restored MICA/B expression was dependent on ATR as well as E2F1, a transcription factor. We further revealed that low‑dose treatment of an HDAC inhibitor was sufficient to restore MICA/B expression in insensitive cells. Finally, we demonstrated that HDAC inhibition restored DNA damage‑dependent cytotoxic NK activity against insensitive cells. Thus, the present study revealed that DNA damage‑dependent MICA/B expression in insensitive cancer cells can be restored by chromatin relaxation via the HDAC/Suv39/G9a pathway. Collectively, manipulation of chromatin status by therapeutic cancer drugs may potentiate the antitumor effect by enhancing immune activation following radiotherapy and DNA damage-associated chemotherapy.

MeSH terms

  • Ataxia Telangiectasia Mutated Proteins / genetics
  • Chromatin Assembly and Disassembly / genetics
  • Cytotoxicity, Immunologic / genetics
  • DNA Damage / genetics
  • E2F1 Transcription Factor / genetics
  • Gene Expression Regulation, Neoplastic
  • Histocompatibility Antigens / genetics
  • Histocompatibility Antigens Class I / genetics*
  • Histocompatibility Antigens Class I / immunology
  • Histone Deacetylases / genetics*
  • Histone Deacetylases / immunology
  • Histone-Lysine N-Methyltransferase / genetics
  • Humans
  • Killer Cells, Natural / immunology
  • Methyltransferases / genetics
  • NK Cell Lectin-Like Receptor Subfamily K / genetics
  • Neoplasms / genetics
  • Neoplasms / immunology*
  • Neoplasms / pathology
  • Repressor Proteins / genetics
  • Tumor Microenvironment / immunology

Substances

  • E2F1 Transcription Factor
  • E2F1 protein, human
  • Histocompatibility Antigens
  • Histocompatibility Antigens Class I
  • KLRK1 protein, human
  • MHC class I-related chain A
  • MICB antigen
  • NK Cell Lectin-Like Receptor Subfamily K
  • Repressor Proteins
  • SUV39H1 protein, human
  • Methyltransferases
  • EHMT2 protein, human
  • Histone-Lysine N-Methyltransferase
  • ATR protein, human
  • Ataxia Telangiectasia Mutated Proteins
  • Histone Deacetylases