Alterations in the glycome after HDAC inhibition impact oncogenic potential in epigenetically plastic SW13 cells

BMC Cancer. 2019 Jan 16;19(1):79. doi: 10.1186/s12885-018-5129-4.

Abstract

Background: Defects in the type and degree of cellular glycosylation impact oncogenesis on multiple levels. Although the type of glycosylation is determined by protein sequence encoded by the genome, the extent and modifications of glycosylation depends on the activity of biosynthetic enzymes and recent data suggests that the glycome is also subject to epigenetic regulation. This study focuses on the ability of HDAC inhibition to alter glycosylation and to lead to pro-oncogenic alterations in the glycome as assessed by metastatic potential and chemoresistance.

Methods: Epigenetically plastic SW13 adrenocortical carcinoma cells were treated with FK228, an HDAC inhibitor with high affinity for HDAC1 and, to a lesser extent, HDAC2. In comparing HDAC inhibitor treated and control cells, differential expression of glycome-related genes were assessed by microarray. Differential glycosylation was then assessed by lectin binding arrays and the ability of cellular proteins to bind to glycans was assessed by glycan binding arrays. Differential sensitivity to paclitaxel, proliferation, and MMP activity were also assessed.

Results: Treatment with FK228 alters expression of enzymes in the biosynthetic pathways for a large number of glycome related genes including enzymes in all major glycosylation pathways and several glycan binding proteins. 84% of these differentially expressed glycome-related genes are linked to cancer, some as prognostic markers and others contributing basic oncogenic functions such as metastasis or chemoresistance. Glycan binding proteins also appear to be differentially expressed as protein extracts from treated and untreated cells show differential binding to glycan arrays. The impact of differential mRNA expression of glycosylation enzymes was documented by differential lectin binding. However, the assessment of changes in the glycome is complicated by the fact that detection of differential glycosylation through lectin binding is dependent on the methods used to prepare samples as protein-rich lysates show different binding than fixed cells in several cases. Paralleling the alterations in the glycome, treatment of SW13 cells with FK228 increases metastatic potential and reduces sensitivity to paclitaxel.

Conclusions: The glycome is substantially altered by HDAC inhibition and these changes may have far-reaching impacts on oncogenesis.

Keywords: Chemoresistance; Epigenetics; Gene expression profiling; Glycan Array; Glycome expression; HDAC inhibition; Lectin Array.

MeSH terms

  • Antineoplastic Agents, Phytogenic / therapeutic use
  • Biosynthetic Pathways / drug effects
  • Cell Line, Tumor
  • Cell Plasticity / drug effects*
  • Cell Plasticity / genetics
  • Cell Transformation, Neoplastic / chemically induced*
  • Cell Transformation, Neoplastic / genetics
  • Depsipeptides / pharmacology
  • Drug Resistance, Neoplasm / drug effects
  • Drug Resistance, Neoplasm / genetics
  • Epigenesis, Genetic / drug effects
  • Glycosylation / drug effects
  • Histone Deacetylase 1 / antagonists & inhibitors
  • Histone Deacetylase 1 / metabolism
  • Histone Deacetylase 2 / antagonists & inhibitors
  • Histone Deacetylase 2 / metabolism
  • Histone Deacetylase Inhibitors / adverse effects*
  • Humans
  • Neoplasms / drug therapy*
  • Neoplasms / genetics
  • Neoplasms / pathology
  • Paclitaxel / pharmacology
  • Paclitaxel / therapeutic use
  • Polysaccharides / metabolism
  • Protein Processing, Post-Translational / drug effects
  • Protein Processing, Post-Translational / genetics

Substances

  • Antineoplastic Agents, Phytogenic
  • Depsipeptides
  • Histone Deacetylase Inhibitors
  • Polysaccharides
  • romidepsin
  • HDAC1 protein, human
  • HDAC2 protein, human
  • Histone Deacetylase 1
  • Histone Deacetylase 2
  • Paclitaxel