Histone variant H3F3A promotes lung cancer cell migration through intronic regulation

Nat Commun. 2016 Oct 3:7:12914. doi: 10.1038/ncomms12914.

Abstract

Although several somatic single nucleotide variations in histone H3.3 have been investigated as cancer drivers, other types of aberration have not been well studied. Here, we demonstrate that overexpression of H3F3A, encoding H3.3, is associated with lung cancer progression and promotes lung cancer cell migration by activating metastasis-related genes. H3.3 globally activates gene expression through the occupation of intronic regions in lung cancer cells. Moreover, H3.3 binding regions show characteristics of regulatory DNA elements. We show that H3.3 is deposited at a specific intronic region of GPR87, where it modifies the chromatin status and directly activates GPR87 transcription. The expression levels of H3F3A and GPR87, either alone or in combination, are robust prognostic markers for early-stage lung cancer, and may indicate potential for the development of treatments involving GPR87 antagonists. In summary, our results demonstrate that intronic regulation by H3F3A may be a target for the development of novel therapeutic strategies.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Carcinoma, Non-Small-Cell Lung / metabolism*
  • Carcinoma, Non-Small-Cell Lung / pathology
  • Cell Line, Tumor
  • Cell Movement
  • Chromatin / chemistry
  • Disease Progression
  • Gene Dosage
  • Gene Expression Profiling
  • Gene Expression Regulation, Neoplastic
  • Histones / metabolism*
  • Humans
  • Introns*
  • Lung Neoplasms / metabolism*
  • Lung Neoplasms / pathology
  • Mutation
  • Neoplasm Metastasis
  • Prognosis
  • Proportional Hazards Models
  • Receptors, Lysophosphatidic Acid / metabolism
  • Regulatory Elements, Transcriptional

Substances

  • Chromatin
  • GPR87 protein, human
  • Histones
  • Receptors, Lysophosphatidic Acid