Assessment of p.Phe508del-CFTR functional restoration in pediatric primary cystic fibrosis airway epithelial cells

PLoS One. 2018 Jan 23;13(1):e0191618. doi: 10.1371/journal.pone.0191618. eCollection 2018.

Abstract

Background: Mutations in the cystic fibrosis transmembrane regulator (CFTR) gene can reduce function of the CFTR ion channel activity and impair cellular chloride secretion. The gold standard method to assess CFTR function of ion transport using the Ussing chamber requires a high number of airway epithelial cells grown at air-liquid interface, limiting the application of this method for high throughput screening of potential therapeutic compounds in primary airway epithelial cells (pAECs) featuring less common CFTR mutations. This study assessed an alternative approach, using a small scale halide assay that can be adapted for a personalized high throughput setting to analyze CFTR function of pAEC.

Methods: Pediatric pAECs derived from children with CF (pAECCF) were established and expanded as monolayer cultures, before seeding into 96-well plates for the halide assay. Cells were then transduced with an adenoviral construct containing yellow fluorescent protein (eYFP) reporter gene, alone or in combination with either wild-type CFTR (WT-CFTR) or p.Phe508del CFTR. Four days post transduction, cells were stimulated with forskolin and genistein, and assessed for quenching of the eYFP signal following injection of iodide solution into the assay media.

Results: Data showed that pAECCF can express eYFP at high efficiency following transduction with the eYFP construct. The halide assay was able to discriminate functional restoration of CFTR in pAECCF treated with either WT-CFTR construct or the positive controls syntaxin 8 and B-cell receptor-associated protein 31 shRNAs.

Significance: The current study demonstrates that the halide assay can be adapted for pediatric pAECCF to evaluate restoration of CFTR function. With the ongoing development of small molecules to modulate the folding and/or activity of various mutated CFTR proteins, this halide assay presents a small-scale personalized screening platform that could assess therapeutic potential of molecules across a broad range of CFTR mutations.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adenoviridae / genetics
  • Bronchi / cytology
  • Bronchi / metabolism*
  • Cells, Cultured
  • Child
  • Cystic Fibrosis / pathology
  • Cystic Fibrosis / physiopathology*
  • Cystic Fibrosis Transmembrane Conductance Regulator / chemistry
  • Cystic Fibrosis Transmembrane Conductance Regulator / genetics
  • Cystic Fibrosis Transmembrane Conductance Regulator / physiology*
  • Epithelial Cells / metabolism
  • Genetic Vectors
  • Humans
  • Phenylalanine / chemistry*
  • Protein Transport
  • Trachea / cytology
  • Trachea / metabolism*
  • Transduction, Genetic

Substances

  • Cystic Fibrosis Transmembrane Conductance Regulator
  • Phenylalanine

Grants and funding

This work was supported by a grant from the Australian Cystic Fibrosis Research Trust, National Health and Medical Research Council of Australia (303145; 458513, 513730) and Cystic Fibrosis Foundation Therapeutics (SLY04A0, STICK09A0). Michela A. Tessari and Richard A. Janssen are employees of Galapagos BV, and David F. Fischer is from Charles River Laboratories. Both Galapagos BV and Charles River Laboratories provided research materials and financial support to the relevant authors (MA Tessari, RA Janssen and DF Fischer) and did not play a role in the study design, data collection and analysis, decision to publish and the preparation of the manuscript. Those authors were involved in reviewing the manuscript prior to submission. Stephen M. Stick is a National Health and Medical Research Council of Australia Practitioner Fellow. The other funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.