Cystine/Glutamate Antiporter (xCT) Is Required for Chief Cell Plasticity After Gastric Injury

Cell Mol Gastroenterol Hepatol. 2019;8(3):379-405. doi: 10.1016/j.jcmgh.2019.04.015. Epub 2019 May 6.

Abstract

Background & aims: Many differentiated epithelial cell types are able to reprogram in response to tissue damage. Although reprogramming represents an important physiological response to injury, the regulation of cellular plasticity is not well understood. Damage to the gastric epithelium initiates reprogramming of zymogenic chief cells into a metaplastic cell lineage known as spasmolytic polypeptide-expressing metaplasia (SPEM). The present study seeks to identify the role of xCT, a cystine/glutamate antiporter, in chief cell reprogramming after gastric injury. We hypothesize that xCT-dependent reactive oxygen species (ROS) detoxification is required for the reprogramming of chief cells into SPEM.

Methods: Sulfasalazine (an xCT inhibitor) and small interfering RNA knockdown were used to target xCT on metaplastic cells in vitro. Sulfasalazine-treated wild-type mice and xCT knockout mice were analyzed. L635 or DMP-777 treatment was used to chemically induce acute gastric damage. The anti-inflammatory metabolites of sulfasalazine (sulfapyridine and mesalazine) were used as controls. Normal gastric lineages, metaplastic markers, autophagy, proliferation, xCT activity, ROS, and apoptosis were assessed.

Results: xCT was up-regulated early as chief cells transitioned into SPEM. Inhibition of xCT or small interfering RNA knockdown blocked cystine uptake and decreased glutathione production by metaplastic cells and prevented ROS detoxification and proliferation. Moreover, xCT activity was required for chief cell reprogramming into SPEM after gastric injury in vivo. Chief cells from xCT-deficient mice showed decreased autophagy, mucus granule formation and proliferation, as well as increased levels of ROS and apoptosis compared with wild-type mice. On the other hand, the anti-inflammatory metabolites of sulfasalazine did not affect SPEM development.

Conclusions: The results presented here suggest that maintaining redox balance is crucial for progression through the reprogramming process and that xCT-mediated cystine uptake is required for chief cell plasticity and ROS detoxification.

Keywords: Autophagy; CD44; Cellular Plasticity; Chief Cell; Metaplasia; Oxyntic Atrophy; Reactive Oxygen Species; Reprogramming; SPEM; Sulfasalazine; xCT.

Publication types

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

MeSH terms

  • Amino Acid Transport System y+ / genetics*
  • Amino Acid Transport System y+ / metabolism
  • Animals
  • Azetidines / adverse effects*
  • Cell Line
  • Cell Plasticity
  • Cellular Reprogramming
  • Chief Cells, Gastric / cytology
  • Chief Cells, Gastric / drug effects
  • Chief Cells, Gastric / metabolism
  • Gastric Mucosa / cytology
  • Gastric Mucosa / drug effects
  • Gastric Mucosa / metabolism
  • Gastric Mucosa / pathology*
  • Gene Knockout Techniques
  • Humans
  • Mice
  • Parietal Cells, Gastric / cytology
  • Parietal Cells, Gastric / drug effects
  • Parietal Cells, Gastric / metabolism
  • Piperazines / adverse effects*
  • Reactive Oxygen Species / metabolism
  • Sulfasalazine / pharmacology*
  • Up-Regulation

Substances

  • Amino Acid Transport System y+
  • Azetidines
  • Piperazines
  • Reactive Oxygen Species
  • SLC7A11 protein, human
  • Slc7a11 protein, mouse
  • DMP 777
  • Sulfasalazine