Gastroenterology

Gastroenterology

Volume 122, Issue 1, January 2002, Pages 119-133
Gastroenterology

Basic Research
Genetic or chemical hypochlorhydria is associated with inflammation that modulates parietal and G-cell populations in mice,**,

https://doi.org/10.1053/gast.2002.30298Get rights and content

Abstract

Background & Aims: Reduced gastric acid predisposes the stomach to colonization by bacteria and inflammation. Therefore, we investigated how the chronic gastritis in mice made hypochlorhydric by either gastrin deficiency or omeprazole treatment modulates epithelial cell function. Methods: The gastric pathology of 16-week-old wild-type gastrin-expressing (G+/+) and gastrin-deficient (G−/−) mice maintained in conventional housing was compared. G−/− mice were then treated with antibiotics for 20 days. In a separate experiment, G+/+ mice were treated with omeprazole for 2 months or treated with omeprazole and antibiotics. Results: Compared with the G+/+ animals, the hypochlorhydric G−/− mice showed significant inflammation that resolved after 20 days of antibiotic treatment and correlated with a decrease in bacterial overgrowth. Elevated G- and parietal-cell numbers in the G−/− mice, quantified by flow cytometry, normalized after antibiotic treatment. G+/+ mice treated with omeprazole had increased bacteria and mucosal lymphocytes that resolved after antibiotic therapy. Quantitation of the gastric cells in these omeprazole-treated mice revealed a significant increase in G- and parietal-cell numbers. On resolution of the gastritis, a decrease in parietal and gastrin-expressing (G) cells was observed despite sustained hypochlorhydria in the presence of omeprazole. Conclusions: Genetic or chemical hypochlorhydria predisposes the stomach to bacterial overgrowth resulting in inflammation. The specific changes in parietal and G cells correlate with the presence of inflammation and not directly with gastric acid. Thus, the normal stomach responds to inflammation by increasing the number and function of cell types that are able to maximize gastric acid output.

GASTROENTEROLOGY 2002;122:119-133

Section snippets

Animals

G−/− (n = 12) mice and strain-matched (129/Sv) wild-type controls (gastrin-expressing [G+/+], n = 12) were bred by homozygous mating. Mice were kept in individual, sterile, microisolator cages in nonbarrier mouse rooms for 16 weeks (conventional housing). Food and water were not autoclaved. No additional procedures were used to house the mice in rooms separate from those that have been intentionally infected with other bacterial pathogens. All mice were fasted overnight with access to water ad

Mucosal inflammation in G−/− mice

The effect of hypochlorhydria on gastric pathology in the G−/− mice was evaluated. Sixteen-week-old G−/− mice showed mucosal thickening and significant inflammation (Figure 1B) compared with the G+/+ mice (Figure 1A).

. Representative H&E and PAS/alcian blue stains from mouse stomachs. Representative H&E-stained sections of inflamed stomach from a 16-week-old (A) G+/+ and (B) G−/− mouse with severe inflammation (arrow) and (C) metaplasia (open arrow), (D) neutral (red arrowhead) and acid (black

Discussion

The findings reported here show that a major consequence of hypochlorhydria induced genetically or chemically is the generation of an inflammatory response. In G−/− mice, lymphocytes were elevated above levels measured in the wild-type (G+/+) animals. Similarly, wild-type mice treated with omeprazole had increased numbers of lymphocytes, indicative of chronic inflammation. This result is consistent with prior studies showing that humans treated with omeprazole also become colonized with gastric

Acknowledgements

The authors thank the flow cytometry core in the University of Michigan Cancer Center (CA46952) and University of Michigan Multipurpose Arthritic Center (AR20557), and the assistance of the radioligand core of the University of Michigan Gastrointestinal Peptide Research Center (DK-34533) for their technical assistance. They also acknowledge the use of rabbit gastrin antibody #1296 from CURE DDRC/RIA Core (DK41301). The authors thank Chris Dickinson for synthesizing iodinated gastrin peptide,

References (51)

  • TC Wang et al.

    Synergistic interaction between hypergastrinemia and Helicobacter infection in a mouse model of gastric cancer

    Gastroenterology

    (2000)
  • MJ Blaser et al.

    Parasitism by the “slow” bacterium Helicobacter pylori leads to altered gastric homeostasis and neoplasia

    J Clin Invest

    (1994)
  • DY Graham

    Helicobacter pylori infection is the primary cause of gastric cancer

    J Gastroenterol

    (2000)
  • A Nomura et al.

    Helicobacter pylori infection and gastric carcinoma among Japanese Americans in Hawaii

    N Engl J Med

    (1991)
  • KA Roth et al.

    Cellular immune responses are essential for the development of Helicobacter felis-associated gastric pathology

    J Immunol

    (1999)
  • LE Smythies et al.

    Helicobacter pylori-induced mucosal inflammation is Th1 mediated and exacerbated in IL-4, but not IFN-gamma, gene-deficient mice

    J Immunol

    (2000)
  • M Sumii et al.

    Expression of antral gastrin and somatostatin mRNA in Helicobacter pylori-infected subjects

    Am J Gastroenterol

    (1994)
  • JG Fox et al.

    High-salt diet induces gastric epithelial hyperplasia and parietal cell loss, and enhances Helicobacter pylori colonization in C57BL/6 mice

    Cancer Res

    (1999)
  • J Parsonnet et al.

    Helicobacter pylori infection and the risk of gastric carcinoma

    N Engl J Med

    (1991)
  • D Jonkers et al.

    Helicobacter pylori and non-Helicobacter pylori bacterial flora in gastric mucosal and tumour specimens of patients with primary gastric lymphoma

    Eur J Clin Invest

    (1997)
  • D Palli

    Epidemiology of gastric cancer: an evaluation of available evidence

    J Gastroenterol

    (2000)
  • RW Stockbruegger et al.

    Pernicious anaemia, intragastric bacterial overgrowth, and possible consequences

    Scand J Gastroenterol

    (1984)
  • RI Walker et al.

    Intestinal barriers to bacteria and their toxins

    Annu Rev Med

    (1990)
  • IA Gisbertz et al.

    Specific detection of Helicobacter pylori and non-Helicobacter pylori flora in small- and large-cell primary gastric B-cell non-Hodgkin's lymphoma

    Ann Oncol

    (1997)
  • D Jonkers et al.

    Differential features of gastric cancer patients, either Helicobacter pylori positive or Helicobacter pylori negative

    Ital J Gastroenterol Hepatol

    (1999)
  • Cited by (110)

    • Hormonal Suppression of Stem Cells Inhibits Symmetric Cell Division and Gastric Tumorigenesis

      2020, Cell Stem Cell
      Citation Excerpt :

      We show here that gastrin deficiency or carcinogen exposure leads to increased symmetric cell division, accumulation of mutations, and cancer progression. While hypergastrinemia promotes gastric corpus proliferation and tumorigenesis (Hayakawa et al., 2016), gastrin deficiency has been linked to antral tumor development (Zavros et al., 2002). We have shown that gastrin-deficient mice are more susceptible to MNU-induced antral tumorigenesis and that elevations in amidated gastrin suppressed antral tumorigenesis in a manner that correlated with gene silencing and epigenetic alterations of Tff1 (Takaishi et al., 2009; Tomita et al., 2011).

    • Clinical Manifestations of Helicobacter pylori–Negative Gastritis

      2017, Clinical Gastroenterology and Hepatology
    • Gastrin and upper GI cancers

      2016, Current Opinion in Pharmacology
      Citation Excerpt :

      Similarly, in studies on Mongolian gerbils infected with H. pylori, long-term PPI administration promotes the progression of atrophic corpus gastritis and cancer [26]. However, reports emerged soon thereafter that gastrin knockout (GAS-KO) mice also developed tumors of the stomach, often spontaneously [27–29]. GAS-KO mice showed hypochlorhydria and bacterial overgrowth, and tumor development was promoted by conventional housing, suggesting the role of abnormal bacterial colonization and subsequent inflammation induced by hypochrorhydria.

    View all citing articles on Scopus

    Address requests for reprints to: Juanita L. Merchant, M.D., Ph.D., 1150 West Medical Drive, MSRB I, 3510, Ann Arbor, Michigan 48109-0650. e-mail: [email protected]; fax: (734) 936-1400.

    **

    Supported in part by Public Health Service grant DK-45729 (to J.L.M.) and DK48815 (to L.C.S.).

    Y.Z. is an associate and J.L.M. is an assistant investigator of the Howard Hughes Medical Institute.

    View full text