Dual oxidase-1 is required for airway epithelial cell migration and bronchiolar reepithelialization after injury

Am J Respir Cell Mol Biol. 2013 Mar;48(3):337-45. doi: 10.1165/rcmb.2012-0393OC. Epub 2012 Dec 13.

Abstract

The respiratory epithelium plays a critical role in innate defenses against airborne pathogens and pollutants, and alterations in epithelial homeostasis and repair mechanisms are thought to contribute to chronic lung diseases associated with airway remodeling. Previous studies implicated the nicotinamide adenine dinucleotide phosphate-reduced oxidase dual oxidase-1 (DUOX1) in redox signaling pathways involved in in vitro epithelial wound responses to infection and injury. However, the importance of epithelial DUOX1 in in vivo epithelial repair pathways has not been established. Using small interfering (si)RNA silencing of DUOX1 expression, we show the critical importance of DUOX1 in wound responses in murine tracheal epithelial (MTE) cells in vitro, as well as its contribution to epithelial regeneration in vivo in a murine model of epithelial injury induced by naphthalene, a selective toxicant of nonciliated respiratory epithelial cells (club cells [Clara]). Whereas naphthalene-induced club-cell injury is normally followed by epithelial regeneration after 7 and 14 days, such airway reepithelialization was significantly delayed after the silencing of airway DUOX1 by oropharyngeal administration of DUOX1-targeted siRNA. Wound closure in MTE cells was related to DUOX1-dependent activation of the epidermal growth factor receptor (EGFR) and the transcription factor signal transducer and activator of transcription-3 (STAT3), known mediators of epithelial cell migration and wound responses. Moreover, in vivo DUOX1 silencing significantly suppressed naphthalene-induced activation of STAT3 and EGFR during early stages of epithelial repair. In conclusion, these experiments demonstrate for the first time an important function for epithelial DUOX1 in lung epithelial regeneration in vivo, by promoting EGFR-STAT3 signaling and cell migration as critical events in initial repair.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Bronchioles / cytology
  • Bronchioles / enzymology
  • Bronchioles / physiology*
  • Cell Movement / physiology*
  • Cells, Cultured
  • Dual Oxidases
  • Epithelial Cells / cytology
  • Epithelial Cells / enzymology
  • Epithelial Cells / metabolism
  • Epithelial Cells / physiology*
  • ErbB Receptors / metabolism
  • Mice
  • Mice, Inbred C57BL
  • NADPH Oxidases / metabolism*
  • Re-Epithelialization / physiology*
  • Respiratory Mucosa / cytology
  • Respiratory Mucosa / enzymology
  • Respiratory Mucosa / metabolism
  • Respiratory Mucosa / physiology*
  • STAT3 Transcription Factor / metabolism
  • Signal Transduction / physiology
  • Trachea / metabolism
  • Wound Healing / physiology*

Substances

  • STAT3 Transcription Factor
  • Stat3 protein, mouse
  • Dual Oxidases
  • NADPH Oxidases
  • Duox1 protein, mouse
  • EGFR protein, mouse
  • ErbB Receptors