The role of epigenetic modification in tumorigenesis and progression of pituitary adenomas: a systematic review of the literature

PLoS One. 2013 Dec 18;8(12):e82619. doi: 10.1371/journal.pone.0082619. eCollection 2013.

Abstract

Background: Pituitary adenomas (PAs) are commonly occurring neoplasms with diverse endocrine and neurological effects. Although somatic gene mutations are uncommon in sporadic PAs, recent studies lend support to epigenetic modification as a potential cause of tumorigenesis and tumor progression.

Methods: A systematic literature review of the PubMed and Google Scholar databases was conducted to identify abstracts (n=1,082) pertaining to key targets and mechanisms implicated in epigenetic dysregulation of PAs published between 1993-2013. Data regarding histopathological subtype, target genes, mode of epigenetic modification, and clinical correlation were recorded and analyzed.

Results: Of the 47 that studies met inclusion criteria and focused on epigenomic assessment of PAs, only 2 were genome-scale analyses. Current evidence supports epigenetic alteration in at least 24 PA genes, which were categorized into four groups based on function and epigenetic alteration: 1) Sixteen tumor suppressor genes silenced via DNA methylation; 2) Two oncogenes overexpressed via histone acetylation and hypomethylation; 3) Three imprinted genes with selective allelic silencing; and 4) One epigenome writer inducing abnormal genome-scale activity and 5) Two transcription regulators indirectly modifying the genome. Of these, 5 genes (CDKN2A, GADD45y, FGFR2, caspase-8, and PTAG) showed particular susceptibility to epigenetic modification, with abnormal DNA methylation in >50% of PA samples. Several genes displayed correlations between epigenetic modification and clinically relevant parameters, including invasiveness (CDKN2A; DAPK; Rb1), sex (MAGE-A3), tumor size (GNAS1), and histopathological subtype (CDKN2A; MEG3; p27; RASSF1A; Rb1).

Conclusions: Epigenetic modification of selected PA genes may play a key role in tumorigenesis and progression, which may translate into important diagnostic and therapeutic applications.

Publication types

  • Review
  • Systematic Review

MeSH terms

  • Apoptosis Regulatory Proteins / genetics
  • Caspase 8 / genetics
  • Cyclin-Dependent Kinase Inhibitor p16 / genetics
  • DNA Methylation / genetics
  • Epigenesis, Genetic / genetics*
  • Humans
  • Pituitary Neoplasms / genetics*
  • Receptor, Fibroblast Growth Factor, Type 2 / genetics

Substances

  • Apoptosis Regulatory Proteins
  • Cyclin-Dependent Kinase Inhibitor p16
  • RHBDD3 protein, human
  • FGFR2 protein, human
  • Receptor, Fibroblast Growth Factor, Type 2
  • Caspase 8