Characterization of the Potent, Selective Nrf2 Activator, 3-(Pyridin-3-Ylsulfonyl)-5-(Trifluoromethyl)-2 H-Chromen-2-One, in Cellular and In Vivo Models of Pulmonary Oxidative Stress

J Pharmacol Exp Ther. 2017 Oct;363(1):114-125. doi: 10.1124/jpet.117.241794. Epub 2017 Aug 8.

Abstract

Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a key regulator of oxidative stress and cellular repair and can be activated through inhibition of its cytoplasmic repressor, Kelch-like ECH-associated protein 1 (Keap1). Several small molecule disrupters of the Nrf2-Keap1 complex have recently been tested and/or approved for human therapeutic use but lack either potency or selectivity. The main goal of our work was to develop a potent, selective activator of NRF2 as protection against oxidative stress. In human bronchial epithelial cells, our Nrf2 activator, 3-(pyridin-3-ylsulfonyl)-5-(trifluoromethyl)-2H-chromen-2-one (PSTC), induced Nrf2 nuclear translocation, Nrf2-regulated gene expression, and downstream signaling events, including induction of NAD(P)H:quinone oxidoreductase 1 (NQO1) enzyme activity and heme oxygenase-1 protein expression, in an Nrf2-dependent manner. As a marker of subsequent functional activity, PSTC restored oxidant (tert-butyl hydroperoxide)-induced glutathione depletion. The compound's engagement of the Nrf2 signaling pathway translated to an in vivo setting, with induction of Nrf2-regulated gene expression and NQO1 enzyme activity, as well as restoration of oxidant (ozone)-induced glutathione depletion, occurring in the lungs of PSTC-treated rodents. Under disease conditions, PSTC engaged its target, inducing the expression of Nrf2-regulated genes in human bronchial epithelial cells derived from patients with chronic obstructive pulmonary disease, as well as in the lungs of cigarette smoke-exposed mice. Subsequent to the latter, a dose-dependent inhibition of cigarette smoke-induced pulmonary inflammation was observed. Finally, in contrast with bardoxolone methyl and sulforaphane, PSTC did not inhibit interleukin-1β-induced nuclear factor-κB translocation or insulin-induced S6 phosphorylation in human cells, emphasizing the on-target activity of this compound. In summary, we characterize a potent, selective Nrf2 activator that offers protection against pulmonary oxidative stress in several cellular and in vivo models.

MeSH terms

  • Animals
  • Blotting, Western
  • Cell Line
  • Cell Nucleus / metabolism
  • Coumarins / administration & dosage
  • Coumarins / blood
  • Coumarins / therapeutic use*
  • Disease Models, Animal
  • Drug Discovery
  • Epithelial Cells / drug effects*
  • Epithelial Cells / metabolism
  • Gene Expression / drug effects
  • Glutathione / metabolism
  • HEK293 Cells
  • Humans
  • Lung / drug effects*
  • Lung / metabolism
  • Mice, Inbred C57BL
  • NAD(P)H Dehydrogenase (Quinone) / genetics
  • NF-E2-Related Factor 2 / agonists*
  • NF-E2-Related Factor 2 / genetics
  • Oxidative Stress / drug effects*
  • Ozone / toxicity
  • Pneumonia / etiology
  • Pneumonia / metabolism
  • Pneumonia / prevention & control*
  • Protein Transport
  • Pulmonary Disease, Chronic Obstructive / metabolism*
  • RNA, Small Interfering / genetics
  • Rats, Wistar
  • Smoking / adverse effects
  • Sulfones / administration & dosage
  • Sulfones / blood
  • Sulfones / therapeutic use*
  • Transfection

Substances

  • 3-(pyridin-3-ylsulfonyl)-5-(trifluoromethyl)-2H-chromen-2-one
  • Coumarins
  • NF-E2-Related Factor 2
  • NFE2L2 protein, human
  • RNA, Small Interfering
  • Sulfones
  • Ozone
  • NAD(P)H Dehydrogenase (Quinone)
  • Glutathione