Improved Homology Model of the Human all-trans Retinoic Acid Metabolizing Enzyme CYP26A1

Molecules. 2016 Mar 15;21(3):351. doi: 10.3390/molecules21030351.

Abstract

A new CYP26A1 homology model was built based on the crystal structure of cyanobacterial CYP120A1. The model quality was examined for stereochemical accuracy, folding reliability, and absolute quality using a variety of different bioinformatics tools. Furthermore, the docking capabilities of the model were assessed by docking of the natural substrate all-trans-retinoic acid (atRA), and a group of known azole- and tetralone-based CYP26A1 inhibitors. The preferred binding pose of atRA suggests the (4S)-OH-atRA metabolite production, in agreement with recently available experimental data. The distances between the ligands and the heme group iron of the enzyme are in agreement with corresponding distances obtained for substrates and azole inhibitors for other cytochrome systems. The calculated theoretical binding energies agree with recently reported experimental data and show that the model is capable of discriminating between natural substrate, strong inhibitors (R116010 and R115866), and weak inhibitors (liarozole, fluconazole, tetralone derivatives).

Keywords: CYP26A1; RAMBA; homology model; molecular docking; retinoic acid.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Catalytic Domain
  • Heme / chemistry
  • Humans
  • Hydrogen Bonding
  • Models, Molecular*
  • Molecular Conformation
  • Molecular Docking Simulation
  • Molecular Dynamics Simulation
  • Norisoprenoids / chemistry
  • Protein Binding
  • Retinoic Acid 4-Hydroxylase / chemistry*
  • Tetralones / chemistry
  • Tretinoin / chemistry*

Substances

  • Norisoprenoids
  • Tetralones
  • Heme
  • Tretinoin
  • Retinoic Acid 4-Hydroxylase