Vacuolating cytotoxin and variants in Atg16L1 that disrupt autophagy promote Helicobacter pylori infection in humans

Deepa Raju, Seamus Hussey, Michelle Ang, Mauricio R. Terebiznik, Michael Sibony, Esther Galindo-Mata, Vijay Gupta, Steven R. Blanke, Alberto Delgado, Judith Romero-Gallo, Mahendra Singh Ramjeet, Heidi Mascarenhas, Richard M. Peek, Pelayo Correa, Cathy Streutker, Georgina Hold, Erdmutte Kunstmann, Tamotsu Yoshimori, Mark S. Silverberg, Stephen E. GirardinDana J. Philpott, Emad El Omar, Nicola L. Jones

Research output: Contribution to journalArticlepeer-review

179 Citations (Scopus)
5 Downloads (Pure)

Abstract

BACKGROUND & AIMS: The Helicobacter pylori toxin vacuolating cytotoxin (VacA) promotes gastric colonization and its presence (VacA+) is associated with more-severe disease. The exact mechanisms by which VacA contributes to infection are unclear. We previously found that limited exposure to VacA induces autophagy of gastric cells, which eliminates the toxin; we investigated whether autophagy serves as a defense mechanism against H pylori infection.

METHODS: We investigated the effect of VacA on autophagy in human gastric epithelial cells (AGS) and primary gastric cells from mice. Expression of p62, a marker of autophagy, was also assessed in gastric tissues from patients infected with toxigenic (VacA+) or nontoxigenic strains. We analyzed the effect of VacA on autophagy in peripheral blood monocytes obtained from subjects with different genotypes of ATG16L1, which regulates autophagy. We performed genotyping for ATG16L1in two cohorts of infected and uninfected subjects.

RESULTS: Prolonged exposure of AGS and mouse gastric cells to VacA disrupted induction of autophagy in response to the toxin, because the cells lacked cathepsin-D in autophagosomes. Loss of autophagy resulted in the accumulation of p62 and reactive oxygen species. Gastric biopsies samples from patients infected with VacA+, but not nontoxigenic strains of H pylori, had increased levels of p62. Peripheral blood monocytes isolated from individuals with polymorphisms in ATG16L1that increase susceptibility to Crohn's disease had reduced induction of autophagy in response to VacA+ compared to cells from individuals that did not have these polymorphisms. The presence of the ATG16L1Crohn's disease risk variant increased susceptibility to H pylori infection in 2 separate cohorts.

CONCLUSIONS: Autophagy protects against infection with H pylori; the toxin VacA disrupts autophagy to promote infection, which could contribute to inflammation and eventual carcinogenesis.
Original languageEnglish
Pages (from-to)1160-1171
Number of pages12
JournalGastroenterology
Volume142
Issue number5
Early online date13 Feb 2012
DOIs
Publication statusPublished - May 2012

Keywords

  • stomach cancer
  • genetic
  • bacteria toxin
  • tumor

Fingerprint

Dive into the research topics of 'Vacuolating cytotoxin and variants in Atg16L1 that disrupt autophagy promote Helicobacter pylori infection in humans'. Together they form a unique fingerprint.

Cite this