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)-beta1 negatively regulates Th1 cell development, and TGF-beta1-deficient mice spontaneously develop gastritis. Here, we examined TGF-beta1 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-beta1 by ELISA and Smad3 and 7 expression by Western blotting. Induction of Smad7 by interferon (IFN)-gamma 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-beta1 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-beta1-associated Smad3 phosphorylation, which was associated with high expression of the inhibitor Smad7. Blocking Smad7 with antisense oligonucleotides restored TGF-beta1 signaling in biopsy specimens from Hp-infected patients and concomitantly reduced interferon-gamma and T-bet. Smad7 was inducible in normal gastric biopsy specimens by interferon-gamma through a STAT1-dependent mechanism, and neutralization of interferon-gamma in biopsy specimens from Hp-infected patients reduced Smad7 expression. Conclusions. These data suggest that, in Hp-infected gastric mucosa, interferon-gamma induces the expression of Smad7, which then prevents endogenous TGF-beta1 from down-regulating the ongoing tissue-damaging Th1 response.

Monteleone, G., DEL VECCHIO BLANCO, G., Palmieri, G., Vavassori, P., Monteleone, I., Colantoni, A., et al. (2004). Induction and regulation of Smad7 in the gastric mucosa of patients with Helicobacter pylori infection. GASTROENTEROLOGY, 126(3), 674-682 [10.1053/j.gastro.2003.11.048].

Induction and regulation of Smad7 in the gastric mucosa of patients with Helicobacter pylori infection

MONTELEONE, GIOVANNI;DEL VECCHIO BLANCO, GIOVANNA;PALMIERI, GIAMPIERO;MONTELEONE, IVAN;COLANTONI, ALFREDO;PALLONE, FRANCESCO
2004-01-01

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)-beta1 negatively regulates Th1 cell development, and TGF-beta1-deficient mice spontaneously develop gastritis. Here, we examined TGF-beta1 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-beta1 by ELISA and Smad3 and 7 expression by Western blotting. Induction of Smad7 by interferon (IFN)-gamma 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-beta1 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-beta1-associated Smad3 phosphorylation, which was associated with high expression of the inhibitor Smad7. Blocking Smad7 with antisense oligonucleotides restored TGF-beta1 signaling in biopsy specimens from Hp-infected patients and concomitantly reduced interferon-gamma and T-bet. Smad7 was inducible in normal gastric biopsy specimens by interferon-gamma through a STAT1-dependent mechanism, and neutralization of interferon-gamma in biopsy specimens from Hp-infected patients reduced Smad7 expression. Conclusions. These data suggest that, in Hp-infected gastric mucosa, interferon-gamma induces the expression of Smad7, which then prevents endogenous TGF-beta1 from down-regulating the ongoing tissue-damaging Th1 response.
2004
Pubblicato
Rilevanza internazionale
Articolo
Sì, ma tipo non specificato
Settore MED/12 - GASTROENTEROLOGIA
English
amoxicillin; antisense oligonucleotide; clarithromycin; cytokine; gamma interferon; omeprazole; protein inhibitor; Smad3 protein; Smad7 protein; STAT1 protein; transforming growth factor beta1; adult; aged; article; cell isolation; cell maturation; cytokine production; enzyme linked immunosorbent assay; female; gastritis; gene control; gene induction; Helicobacter infection; Helicobacter pylori; human; major clinical study; male; mucosa cell; pathophysiology; priority journal; protein expression; protein phosphorylation; signal transduction; stomach biopsy; stomach mucosa; Th1 cell; Western blotting; Adult; Aged; DNA-Binding Proteins; Down-Regulation; Enzyme-Linked Immunosorbent Assay; Female; Gastric Mucosa; Gene Expression Regulation; Helicobacter Infections; Helicobacter pylori; Humans; Interferon Type II; Male; Organ Culture Techniques; Phosphorylation; Signal Transduction; Smad3 Protein; Smad7 Protein; T-Box Domain Proteins; Trans-Activators; Transcription Factors; Transforming Growth Factor beta; Transforming Growth Factor beta1
Monteleone, G., DEL VECCHIO BLANCO, G., Palmieri, G., Vavassori, P., Monteleone, I., Colantoni, A., et al. (2004). Induction and regulation of Smad7 in the gastric mucosa of patients with Helicobacter pylori infection. GASTROENTEROLOGY, 126(3), 674-682 [10.1053/j.gastro.2003.11.048].
Monteleone, G; DEL VECCHIO BLANCO, G; Palmieri, G; Vavassori, P; Monteleone, I; Colantoni, A; Battista, S; Spagnoli, L; Romano, M; Borrelli, M; Macdonald, T; Pallone, F
Articolo su rivista
File in questo prodotto:
Non ci sono file associati a questo prodotto.

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/2108/40803
Citazioni
  • ???jsp.display-item.citation.pmc??? 22
  • Scopus 53
  • ???jsp.display-item.citation.isi??? 53
social impact