Gastroenterology

Gastroenterology

Volume 126, Issue 3, March 2004, Pages 674-682
Gastroenterology

Clinical-alimentary tract
Induction and regulation of Smad7 in the gastric mucosa of patients with Helicobacter pylori infection

https://doi.org/10.1053/j.gastro.2003.11.048Get rights and content

Abstract

Background & Aims: Helicobacter pylori (Hp) infection causes a chronic gastric inflammation, which can lead to peptic ulceration and cancer. The inflammatory response is multifactorial and is characterized by exaggerated Th1 cytokine production. How the Th1 response is induced and maintained in the stomach of Hp-infected patients remains unclear. Transforming growth factor (TGF)-β1 negatively regulates Th1 cell development, and TGF-β1-deficient mice spontaneously develop gastritis. Here, we examined TGF-β1 signaling in Hp-associated gastritis. Methods:Gastric biopsy specimens taken from patients with or without Hp infection were analyzed for the content of activated TGF-β1 by ELISA and Smad3 and 7 expression by Western blotting. Induction of Smad7 by interferon (IFN)-γ was examined in normal gastric mucosal biopsy specimens, whereas the effect of Smad7 inhibition on the ongoing Th1 response was analyzed in Hp-colonized biopsy specimens. Results:Activated TGF-β1 was abundant in the mucosa of controls and Hp-infected patients, with no significant difference between the 2 groups. Despite this, in whole biopsy specimens and isolated mucosal cells from Hp-infected patients, there was defective TGF-β1-associated Smad3 phosphorylation, which was associated with high expression of the inhibitor Smad7. Blocking Smad7 with antisense oligonucleotides restored TGF-β1 signaling in biopsy specimens from Hp-infected patients and concomitantly reduced interferon-γ and T-bet. Smad7 was inducible in normal gastric biopsy specimens by interferon-γ through a STAT1-dependent mechanism, and neutralization of interferon-γ in biopsy specimens from Hp-infected patients reduced Smad7 expression. Conclusions:These data suggest that, in Hp-infected gastric mucosa, interferon-γ induces the expression of Smad7, which then prevents endogenous TGF-β1 from down-regulating the ongoing tissue-damaging Th1 response.

Section snippets

Patients and sampling

Sixty-five patients (33 men and 32 women; median age, 49 years; range, 21–74 years) who underwent esophagogastroduodenoscopy for dyspeptic symptoms were studied. Thirty-three patients had Hp infection as confirmed by urease quick test and histology. Five Hp-infected patients were reendoscoped 2 months after cessation of 1-week treatment with omeprazole (20 mg twice daily), amoxycillin (1000 mg twice daily), and clarithromycin (500 mg twice daily). All these 5 patients were cured of Hp infection

Activated TGF-β1 is produced in the human gastric mucosa

TGF-β1 negatively regulates mucosal immune responses, and defects in TGF-β1 production have been associated with the development of gastritis in experimental models.9, 10, 11, 12 Therefore, we first sought to analyze whether the Hp-related gastritis associates with changes in the local concentrations of activated TGF-β1. Because several gastric mucosal cell types have the potential to synthesize TGF-β1, the cytokine was measured in total proteins extracted from whole gastric mucosa rather than

Discussion

In this study, we show that colonization of the stomach with Hp leads to a reduction in Smad3 phosphorylation, a crucial step in the TGF-β1-mediated signal transduction, despite the synthesis of active TGF-β1. The failure in TGF-β1 signaling is associated with high expression of Smad7, an inhibitor of TGF-β1-associated Smad3 phosphorylation. In contrast, abundant phosphorylated Smad3 is seen in the normal gastric mucosa, consistent with constitutively active TGF-β1 signaling.

TGF-β1 is one of

References (26)

  • R. Derynck et al.

    Smadstranscriptional activators of TGF-β responses

    Cell

    (1998)
  • C.M. Horvath et al.

    The state of the STATsrecent developments in the study of signal transduction to the nucleus

    Curr Opin Cell Biol

    (1997)
  • S.J. Szabo et al.

    A novel transcription factor, T-bet, directs Th1 lineage commitment

    Cell

    (2000)
  • P.B. Ernst et al.

    The disease spectrum of Helicobacter pylorithe immunopathogenesis of gastroduodenal ulcer and gastric cancer

    Annu Rev Microbiol

    (2000)
  • E.M. El-Omar et al.

    The role of interleukin-1 polymorphisms in the pathogenesis of gastric cancer

    Nature

    (2000)
  • S.H. Chu et al.

    Role of NF-κB and AP-1 on Helicobacter pylori-induced IL-8 expression in AGS cells

    Dig Dis Sci

    (2003)
  • A. Bhattacharyya et al.

    Mitogen-activated protein kinases and nuclear factor-κB regulate Helicobacter pylori-mediated interleukin-8 release from macrophages

    Biochem J

    (2002)
  • T.L. Cover et al.

    Induction of gastric epithelial cell apoptosis by Helicobacter pylori vacuolating cytotoxin

    Cancer Res

    (2003)
  • M.M. D’Elios et al.

    T helper 1 effector cells specific for Helicobacter pylori in the gastric antrum of patients with peptic ulcer disease

    J Immunol

    (1997)
  • L.E. Smythies et al.

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

    J Immunol

    (2000)
  • K.A. Eaton et al.

    The role of T-cell subsets and cytokines in the pathogenesis of Helicobacter pylori gastritis in mice

    J Immunol

    (2001)
  • J.J. Letterio et al.

    Regulation of immune responses by TGF-β

    Annu Rev Immunol

    (1998)
  • S.M. Wahl

    Transforming growth factor βthe good, the bad, and the ugly

    J Exp Med

    (1994)
  • Cited by (55)

    • SMAD proteins: Mediators of diverse outcomes during infection

      2022, European Journal of Cell Biology
      Citation Excerpt :

      Colonising more than half the global population, H. pylori is linked with gastritis, intestinal metaplasia, and gastric carcinoma (Monteleone et al., 2004; Shuang et al., 2015). H. pylori infection of gastric epithelial cells results in activation of NFκB, secretion of IL-8, and a localised Th1 response with increased expression of IL-12, TNF-α and IFN-γ (Monteleone et al., 2004; Shuang et al., 2015). Since TGF-β is an antagonist to Th1 responses, SMAD7 overexpression is a potential mechanism by which TGF-β signalling is attenuated and its anti-inflammatory effects ablated during H. pylori infection (Fig. 3).

    • The role of transforming growth factor (TGF)-β in modulating the immune response and fibrogenesis in the gut

      2014, Cytokine and Growth Factor Reviews
      Citation Excerpt :

      This is likely to derive from the restoration of endogenous TGF-β activity, as the ex vivo effects of the Smad7 antisense oligonucleotide are decreased by the addition of a TGF-β neutralizing antibody [105]. Elevated levels of Smad7 and impaired TGF-β1 signaling are not specific features of IBD, since the same alterations have been also observed in the gastric mucosa of patients with Helicobacter pylori infection [106]. Treg, while being reduced in peripheral blood of patients with active IBD, are increased in the inflamed intestinal mucosa of IBD patients [107,108].

    • Smad7 antisense oligonucleotide-based therapy for inflammatory bowel diseases

      2013, Digestive and Liver Disease
      Citation Excerpt :

      In line with this was also the observation that treatment of mucosal explants from CD patients with Smad7 antisense oligonucleotide inhibited Smad7 protein, enhanced Smad3 phosphorylation and decreased inflammatory cytokine production, an effect which was partially blocked by a neutralising TGF-β1 antibody [15]. Up-regulation of Smad7 and diminished expression of phosphorylated Smad3 are not however a specific hallmark of IBD, because similar findings were seen in biopsies taken from Helicobacter pylori-infected patients [20]. By contrast, Smad7 is not up-regulated in the duodenum of patients with active celiac disease [21], suggesting that induction of Smad7 is not an epiphenomenon of the ongoing mucosal inflammation.

    • Smad7 protein induces interferon regulatory factor 1-dependent transcriptional activation of caspase 8 to restore tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis

      2013, Journal of Biological Chemistry
      Citation Excerpt :

      Interestingly, halofuginone has recently been shown to inhibit tumor growth and metastasis through the blocking of angiogenesis and matrix metalloproteinase in various tumors (60, 61). It is well known that IFN-γ induces Smad7 through the Jak1/STAT pathway to block TGF-β signaling (30–32). Because cotreatment of IFN-γ and TRAIL synergistically induced the apoptosis in neuroblastoma cells and Ewing sarcoma, it will be interesting to examine the involvement of Smad7 in this effect based on our study (28, 29, 62).

    View all citing articles on Scopus

    Supported by the Broad Medical Research Program Foundation.

    View full text