Pharmacological and molecular dynamics analyses of differences in inhibitor binding to human and nematode PDE4: Implications for management of parasitic nematodes

PLoS One. 2019 Mar 27;14(3):e0214554. doi: 10.1371/journal.pone.0214554. eCollection 2019.

Abstract

Novel chemical controls are needed that selectively target human, animal, and plant parasitic nematodes with reduced adverse effects on the host or the environment. We hypothesize that the phosphodiesterase (PDE) enzyme family represents a potential target for development of novel nematicides and anthelmintics. To test this, we identified six PDE families present in the nematode phylum that are orthologous to six of the eleven human PDE families. We characterized the binding interactions of family-selective PDE inhibitors with human and C. elegans PDE4 in conjunction with molecular dynamics (MD) simulations to evaluate differences in binding interactions of these inhibitors within the PDE4 catalytic domain. We observed that roflumilast (human PDE4-selective inhibitor) and zardaverine (selective for human PDE3 and PDE4) were 159- and 77-fold less potent, respectively, in inhibiting C. elegans PDE4. The pan-specific PDE inhibitor isobutyl methyl xanthine (IBMX) had similar affinity for nematode and human PDE4. Of 32 residues within 5 Å of the ligand binding site, five revealed significant differences in non-bonded interaction energies (van der Waals and electrostatic interaction energies) that could account for the differential binding affinities of roflumilast and zardaverine. One site (Phe506 in the human PDE4D3 amino acid sequence corresponding to Tyr253 in C. elegans PDE4) is predicted to alter the binding conformation of roflumilast and zardaverine (but not IBMX) into a less energetically favorable state for the nematode enzyme. The pharmacological differences in sensitivity to PDE4 inhibitors in conjunction with differences in the amino acids comprising the inhibitor binding sites of human and C. elegans PDE4 catalytic domains together support the feasibility of designing the next generation of anthelmintics/nematicides that could selectively bind to nematode PDEs.

Publication types

  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Amino Acid Sequence
  • Animals
  • Antinematodal Agents / adverse effects
  • Antinematodal Agents / metabolism*
  • Antinematodal Agents / pharmacology*
  • Caenorhabditis elegans / drug effects
  • Caenorhabditis elegans / enzymology*
  • Cyclic Nucleotide Phosphodiesterases, Type 4 / chemistry
  • Cyclic Nucleotide Phosphodiesterases, Type 4 / metabolism*
  • Humans
  • Molecular Dynamics Simulation*
  • Phosphodiesterase 4 Inhibitors / adverse effects
  • Phosphodiesterase 4 Inhibitors / metabolism*
  • Phosphodiesterase 4 Inhibitors / pharmacology*
  • Protein Binding
  • Protein Conformation

Substances

  • Antinematodal Agents
  • Phosphodiesterase 4 Inhibitors
  • Cyclic Nucleotide Phosphodiesterases, Type 4

Grants and funding

This work was supported in part by United State Department of Agriculture-National Institute of Food and Agriculture under award number 2016-67013-24763 (to R.H.C.) and the NH Agricultural Experiment Station Hatch grant NH-00631 (to R.H.C.); this is Scientific Contribution Number 2812. Computations were performed in part on Trillian, a Cray XE6m-200 supercomputer supported by the National Science Foundation (NSF) Major Research Instrumentation program under grant PHY-1229408 (H.V.) and also using the NSF-supported (ACI-1548562) Extreme Science and Engineering Discovery Environment (XSEDE) Comet resource at San Diego Supercomputer Center (SDSC) under grant TG-MCB160183 (to H.V.). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.