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

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 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 ATG16L1 in 2 cohorts of infected and uninfected subjects. RESULTS Prolonged exposure of human gastric epithelial cells 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 biopsy 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 ATG16L1 that 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 ATG16L1 Crohn'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.

[1]  C. Sasakawa,et al.  A Tecpr1-dependent selective autophagy pathway targets bacterial pathogens. , 2011, Cell host & microbe.

[2]  G. Capellá,et al.  Helicobacter pylori cagA and vacA Genotypes as Predictors of Progression of Gastric Preneoplastic Lesions: A Long-Term Follow-Up in a High-Risk Area in Spain , 2011, The American Journal of Gastroenterology.

[3]  G. Bhanot,et al.  Autophagy Suppresses Tumorigenesis through Elimination of p62 , 2011, Cell.

[4]  J. Rubenstein,et al.  Helicobacter pylori DNA decreases pro-inflammatory cytokine production by dendritic cells and attenuates dextran sodium sulphate-induced colitis , 2011, Gut.

[5]  M. Dave,et al.  Association between Helicobacter pylori infection and inflammatory bowel disease: A meta‐analysis and systematic review of the literature† , 2010, Inflammatory bowel diseases.

[6]  R. Peek,et al.  Helicobacter pylori: gastric cancer and beyond , 2010, Nature Reviews Cancer.

[7]  E. White,et al.  Role of autophagy in suppression of inflammation and cancer. , 2010, Current opinion in cell biology.

[8]  R. Xavier,et al.  Crohn's disease‐associated adherent‐invasive E. coli are selectively favoured by impaired autophagy to replicate intracellularly , 2010, Cellular microbiology.

[9]  B. Adams-Huet,et al.  Autophagy genes protect against Salmonella typhimurium infection and mediate insulin signaling-regulated pathogen resistance , 2009, Proceedings of the National Academy of Sciences.

[10]  V. Deretic,et al.  Autophagy, immunity, and microbial adaptations. , 2009, Cell host & microbe.

[11]  M. Diaz-Meco,et al.  p62 at the Crossroads of Autophagy, Apoptosis, and Cancer , 2009, Cell.

[12]  Gyan Bhanot,et al.  Autophagy Suppresses Tumorigenesis through Elimination of p62 , 2009, Cell.

[13]  S. Blanke,et al.  Effect of Helicobacter pylori’s vacuolating cytotoxin on the autophagy pathway in gastric epithelial cells , 2009, Autophagy.

[14]  S. Mousa,et al.  Role of the proteolytic hierarchy between cathepsin L, cathepsin D and caspase-3 in regulation of cellular susceptibility to apoptosis and autophagy. , 2008, Biochimica et biophysica acta.

[15]  S. Akira,et al.  Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1β production , 2008, Nature.

[16]  M. Daly,et al.  Impaired Autophagy of an Intracellular Pathogen Induced by a Crohn's Disease Associated ATG16L1 Variant , 2008, PloS one.

[17]  B. Levine,et al.  Eating the enemy within: autophagy in infectious diseases , 2008, Cell Death and Differentiation.

[18]  Judy H. Cho,et al.  Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease , 2008, Nature Genetics.

[19]  E. El-Omar,et al.  Toxigenic Helicobacter pylori Infection Precedes Gastric Hypochlorhydria in Cancer Relatives, and H. pylori Virulence Evolves in These Families , 2008, Clinical Cancer Research.

[20]  Guido Kroemer,et al.  Autophagy in the Pathogenesis of Disease , 2008, Cell.

[21]  Robin Mathew,et al.  Role of autophagy in cancer , 2007, Nature Reviews Cancer.

[22]  Guido Kroemer,et al.  Self-eating and self-killing: crosstalk between autophagy and apoptosis , 2007, Nature Reviews Molecular Cell Biology.

[23]  T. Noda,et al.  Dissection of the Autophagosome Maturation Process by a Novel Reporter Protein, Tandem Fluorescent-Tagged LC3 , 2007, Autophagy.

[24]  Judy H Cho,et al.  Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis , 2007, Nature Genetics.

[25]  G. Mills,et al.  Silencing mammalian target of rapamycin signaling by small interfering RNA enhances rapamycin-induced autophagy in malignant glioma cells , 2007, Oncogene.

[26]  T. Yoshimori Autophagy: Paying Charon's Toll , 2007, Cell.

[27]  T. P. Neufeld,et al.  Direct Induction of Autophagy by Atg1 Inhibits Cell Growth and Induces Apoptotic Cell Death , 2007, Current Biology.

[28]  T. Wang,et al.  Helicobacter pylori VacA Toxin Promotes Bacterial Intracellular Survival in Gastric Epithelial Cells , 2006, Infection and Immunity.

[29]  R. Maier,et al.  The diverse antioxidant systems of Helicobacter pylori , 2006, Molecular microbiology.

[30]  Hiroshi Sagara,et al.  Escape of Intracellular Shigella from Autophagy , 2005, Science.

[31]  T. Ueno,et al.  LC3 conjugation system in mammalian autophagy , 2004, The International Journal of Biochemistry & Cell Biology.

[32]  M. Colombo,et al.  Rab7 is required for the normal progression of the autophagic pathway in mammalian cells , 2004, Journal of Cell Science.

[33]  K. Kirkegaard,et al.  Cellular autophagy: surrender, avoidance and subversion by microorganisms , 2004, Nature Reviews Microbiology.

[34]  N. Jones,et al.  Helicobacter pylori strains expressing the vacuolating cytotoxin interrupt phagosome maturation in macrophages by recruiting and retaining TACO (coronin 1) protein , 2003, Cellular microbiology.

[35]  S. Falkow,et al.  Helicobacter pylori enter and survive within multivesicular vacuoles of epithelial cells , 2002, Cellular microbiology.

[36]  M. Carrington,et al.  Interleukin-1 polymorphisms associated with increased risk of gastric cancer , 2000, Nature.

[37]  R. Rappuoli,et al.  Effect of Helicobacter pylori Vacuolating Toxin on Maturation and Extracellular Release of Procathepsin D and on Epidermal Growth Factor Degradation* , 1997, The Journal of Biological Chemistry.

[38]  D. Philpott,et al.  Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry , 2010, Nature Immunology.

[39]  M. Washington,et al.  Activation of (cid:1) -catenin by carcinogenic Helicobacter pylori , 2022 .

[40]  Glenn E. Mortimore,et al.  Intracellular Protein Catabolism and its Control During Nutrient Deprivation and Supply , 1987 .