ING1 and 5-azacytidine act synergistically to block breast cancer cell growth

PLoS One. 2012;7(8):e43671. doi: 10.1371/journal.pone.0043671. Epub 2012 Aug 20.

Abstract

Background: Inhibitor of Growth (ING) proteins are epigenetic "readers" that recognize trimethylated lysine 4 of histone H3 (H3K4Me3) and target histone acetyl transferase (HAT) and histone deacetylase (HDAC) complexes to chromatin.

Methods and principal findings: Here we asked whether dysregulating two epigenetic pathways with chemical inhibitors showed synergistic effects on breast cancer cell line killing. We also tested whether ING1 could synergize better with chemotherapeutics that target the same epigenetic mechanism such as the HDAC inhibitor LBH589 (Panobinostat) or a different epigenetic mechanism such as 5-azacytidine (5azaC), which inhibits DNA methyl transferases. Simultaneous treatment of breast cancer cell lines with LBH589 and 5azaC did not show significant synergy in killing cells. However, combination treatment of ING1 with either LBH589 or 5azaC did show synergy. The combination of ING1b with 5azaC, which targets two distinct epigenetic mechanisms, was more effective at lower doses and enhanced apoptosis as determined by Annexin V staining and cleavage of caspase 3 and poly-ADP-ribose polymerase (PARP). ING1b plus 5azaC also acted synergistically to increase γH2AX staining indicating significant levels of DNA damage were induced. Adenoviral delivery of ING1b with 5azaC also inhibited cancer cell growth in a murine xenograft model and led to tumor regression when viral concentration was optimized in vivo.

Conclusions: These data show that targeting distinct epigenetic pathways can be more effective in blocking cancer cell line growth than targeting the same pathway with multiple agents, and that using viral delivery of epigenetic regulators can be more effective in synergizing with a chemical agent than using two chemotherapeutic agents. This study also indicates that the ING1 epigenetic regulator may have additional activities in the cell when expressed at high levels.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antimetabolites, Antineoplastic / therapeutic use
  • Azacitidine / therapeutic use*
  • Breast Neoplasms / drug therapy*
  • Breast Neoplasms / therapy*
  • Cell Line, Tumor
  • Female
  • Humans
  • Hydroxamic Acids / therapeutic use
  • Indoles / therapeutic use
  • Inhibitor of Growth Protein 1
  • Intracellular Signaling Peptides and Proteins / genetics
  • Intracellular Signaling Peptides and Proteins / metabolism*
  • Mice
  • Mice, SCID
  • Nuclear Proteins / genetics
  • Nuclear Proteins / metabolism*
  • Panobinostat
  • Tumor Suppressor Proteins / genetics
  • Tumor Suppressor Proteins / metabolism*
  • Xenograft Model Antitumor Assays

Substances

  • Antimetabolites, Antineoplastic
  • Hydroxamic Acids
  • ING1 protein, human
  • Indoles
  • Inhibitor of Growth Protein 1
  • Intracellular Signaling Peptides and Proteins
  • Nuclear Proteins
  • Tumor Suppressor Proteins
  • Panobinostat
  • Azacitidine