Helicobacter pylori infection impairs chaperone-assisted maturation of Na-K-ATPase in gastric epithelium

Am J Physiol Gastrointest Liver Physiol. 2020 May 1;318(5):G931-G945. doi: 10.1152/ajpgi.00266.2019. Epub 2020 Mar 16.

Abstract

Helicobacter pylori infection always induces gastritis, which may progress to ulcer disease or cancer. The mechanisms underlying mucosal injury by the bacteria are incompletely understood. Here, we identify a novel pathway for H. pylori-induced gastric injury, the impairment of maturation of the essential transport enzyme and cell adhesion molecule, Na-K-ATPase. Na-K-ATPase comprises α- and β-subunits that assemble in the endoplasmic reticulum (ER) before trafficking to the plasma membrane. Attachment of H. pylori to gastric epithelial cells increased Na-K-ATPase ubiquitylation, decreased its surface and total levels, and impaired ion balance. H. pylori did not alter degradation of plasmalemma-resident Na-K-ATPase subunits or their mRNA levels. Infection decreased association of α- and β-subunits with ER chaperone BiP and impaired assembly of α/β-heterodimers, as was revealed by quantitative mass spectrometry and immunoblotting of immunoprecipitated complexes. The total level of BiP was not altered, and the decrease in interaction with BiP was not observed for other BiP client proteins. The H. pylori-induced decrease in Na-K-ATPase was prevented by BiP overexpression, stopping protein synthesis, or inhibiting proteasomal, but not lysosomal, protein degradation. The results indicate that H. pylori impairs chaperone-assisted maturation of newly made Na-K-ATPase subunits in the ER independently of a generalized ER stress and induces their ubiquitylation and proteasomal degradation. The decrease in Na-K-ATPase levels is also seen in vivo in the stomachs of gerbils and chronically infected children. Further understanding of H. pylori-induced Na-K-ATPase degradation will provide insights for protection against advanced disease.NEW & NOTEWORTHY This work provides evidence that Helicobacter pylori decreases levels of Na-K-ATPase, a vital transport enzyme, in gastric epithelia, both in acutely infected cultured cells and in chronically infected patients and animals. The bacteria interfere with BiP-assisted folding of newly-made Na-K-ATPase subunits in the endoplasmic reticulum, accelerating their ubiquitylation and proteasomal degradation and decreasing efficiency of the assembly of native enzyme. Decreased Na-K-ATPase expression contributes to H. pylori-induced gastric injury.

Keywords: Helicobacter pylori; Na-K-ATPase; endoplasmic reticulum; gastric epithelium; protein maturation.

Publication types

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

MeSH terms

  • Cells, Cultured
  • Endoplasmic Reticulum / enzymology*
  • Endoplasmic Reticulum / microbiology
  • Endoplasmic Reticulum Chaperone BiP
  • Enzyme Stability
  • Epithelial Cells / enzymology*
  • Epithelial Cells / microbiology
  • Gastric Mucosa / enzymology*
  • Gastric Mucosa / microbiology
  • Gastritis / enzymology*
  • Gastritis / genetics
  • Gastritis / microbiology
  • Heat-Shock Proteins / metabolism*
  • Helicobacter Infections / enzymology*
  • Helicobacter Infections / genetics
  • Helicobacter Infections / microbiology
  • Helicobacter pylori / pathogenicity*
  • Host-Pathogen Interactions
  • Humans
  • Proteasome Endopeptidase Complex / metabolism
  • Protein Folding
  • Proteolysis
  • Sodium-Potassium-Exchanging ATPase / genetics
  • Sodium-Potassium-Exchanging ATPase / metabolism*
  • Ubiquitination

Substances

  • ATP1B1 protein, human
  • Endoplasmic Reticulum Chaperone BiP
  • Heat-Shock Proteins
  • Proteasome Endopeptidase Complex
  • ATP1A1 protein, human
  • Sodium-Potassium-Exchanging ATPase