Modification of the Gastric Mucosal Microbiota by a Strain-Specific Helicobacter pylori Oncoprotein and Carcinogenic Histologic Phenotype

mBio. 2019 May 28;10(3):e00955-19. doi: 10.1128/mBio.00955-19.

Abstract

Helicobacter pylori is the strongest risk factor for gastric adenocarcinoma; however, most infected individuals never develop this malignancy. Strain-specific microbial factors, such as the oncoprotein CagA, as well as environmental conditions, such as iron deficiency, augment cancer risk. Importantly, dysbiosis of the gastric microbiota is also associated with gastric cancer. To investigate the combinatorial effects of these determinants in an in vivo model of gastric cancer, Mongolian gerbils were infected with the carcinogenic cag+H. pylori strain 7.13 or a 7.13 cagA isogenic mutant, and microbial DNA extracted from gastric tissue was analyzed by 16S rRNA sequencing. Infection with H. pylori significantly increased gastric inflammation and injury, decreased α-diversity, and altered microbial community structure in a cagA-dependent manner. The effect of iron deficiency on gastric microbial communities was also investigated within the context of infection. H. pylori-induced injury was augmented under conditions of iron deficiency, but despite differences in gastric pathology, there were no significant differences in α- or β-diversity, phyla, or operational taxonomic unit (OTU) abundance among infected gerbils maintained on iron-replete or iron-depleted diets. However, when microbial composition was stratified based solely on the severity of histologic injury, significant differences in α- and β-diversity were present among gerbils harboring premalignant or malignant lesions compared to gerbils with gastritis alone. This study demonstrates that H. pylori decreases gastric microbial diversity and community structure in a cagA-dependent manner and that as carcinogenesis progresses, there are corresponding alterations in community structure that parallel the severity of disease.IMPORTANCE Microbial communities are essential for the maintenance of human health, and when these communities are altered, hosts can become susceptible to inflammation and disease. Dysbiosis contributes to gastrointestinal cancers, and specific bacterial species are associated with this phenotype. This study uses a robust and reproducible animal model to demonstrate that H. pylori infection induces gastric dysbiosis in a cagA-dependent manner and further that dysbiosis and altered microbial community structure parallel the severity of H. pylori-induced gastric injury. Ultimately, such models of H. pylori infection and cancer that can effectively evaluate multiple determinants simultaneously may yield effective strategies for manipulating the gastric microbiota to prevent the development of gastric cancer.

Keywords: CagA; Helicobacter pylori; gastric cancer; gastric microbiota; iron deficiency.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Antigens, Bacterial / genetics*
  • Antigens, Bacterial / metabolism
  • Bacterial Proteins / genetics*
  • Bacterial Proteins / metabolism
  • Disease Models, Animal
  • Dysbiosis / microbiology*
  • Gastric Mucosa / microbiology*
  • Gastric Mucosa / pathology
  • Gerbillinae / microbiology
  • Helicobacter Infections / microbiology
  • Helicobacter pylori / genetics
  • Helicobacter pylori / pathogenicity*
  • Male
  • Oncogene Proteins / genetics*
  • Oncogene Proteins / metabolism
  • Phenotype
  • RNA, Ribosomal, 16S / genetics
  • Stomach Neoplasms / microbiology

Substances

  • Antigens, Bacterial
  • Bacterial Proteins
  • Oncogene Proteins
  • RNA, Ribosomal, 16S
  • cagA protein, Helicobacter pylori