Gastroenterology

Gastroenterology

Volume 134, Issue 2, February 2008, Pages 470-480
Gastroenterology

Basic–Alimentary Tract
TNF Receptor Type I-Dependent Activation of Innate Responses to Reduce Intestinal Damage-Associated Mortality

https://doi.org/10.1053/j.gastro.2007.11.055Get rights and content

Background & Aims: Ligation of tumor necrosis factor (TNF) receptors (TNFRs) with TNF plays a critical role in the pathogenesis of human inflammatory bowel disease (IBD). However, it remains unclear which cell types activated through TNFR-associated signaling cascades are involved in the pathogenesis of colitis. Methods: Recombination activating gene-1 (RAG) knockout (KO) (no T or B cells)-based TNFR double and triple KO mice were generated. Bone marrow (BM) chimera mice in which BM-derived myeloid cells, but not colonic epithelial cells (CECs), express TNFRs were also generated. Colitis was induced by administration of dextran sodium sulfate (DSS) in distilled water. Murine lines and chimeras were assessed for disease severity, histopathology, apoptotic cell rate, epithelial proliferation, and bacterial invasion rate. Results: Following DSS administration, mice lacking both RAG and TNFR1 exhibited a high mortality (>80%) rate with an impaired CEC regeneration compared with RAG KO and RAG × TNFR2 double KO (DKO) mice. Transplantation of RAG KO-derived BM cells restored CEC regeneration and rescued the majority of recipient RAG × TNFR1 DKO mice from DSS-induced mortality. After BM transplantation, RAG × TNFR1 DKO mice exhibited an increased rate of apoptosis in the colonic lamina propria macrophages in association with the activation of caspases. In addition, BM reconstitution directly or indirectly enhanced the proliferation of CECs by activating mitogen-activated protein kinase and phosphoinositide-3 kinase/Akt pathways. Conclusions: TNFR1-signaling cascade in colonic myeloid lineage cells contributes to the suppression of acute damage-associated mortality presumably by controlling CEC homeostasis.

Section snippets

Mice and Bone Marrow Transplantation

Six to 8-week-old wild-type (WT), recombination activating gene-1 (RAG) knockout (KO), TNFR1 KO, and TNFR2 KO mice on the C57BL/6 strain (H-2b) were purchased from the Jackson Laboratories (Bar Harbor, ME). To generate RAG KO mice deficient in TNFR1 or TNFR2, RAG KO mice were crossed with TNFR1 KO or TNFR2 KO mice. DNA screening was performed as previously described.13 Briefly, genomic DNA extracted from tail was subject to polymerase chain reaction (PCR). To distinguish heterozygous and

Distinct Effects of TNFR1 vs TNFR2 on Innate Immune Responses Under Acute Intestinal Damage

TNF/TNFR2-mediated signaling on LP T cells is known to play a critical role in the pathogenesis of CD and experimental colitis.19, 20, 21, 22 However, the role of TNFRs in innate immune responses involved in the pathogenesis of colitis has not fully been defined. To address this question, we generated TNFR1 or TNFR2-deficient RAG double KO (DKO) mice that lack T or B cells and induced acute intestinal inflammation by the administration of 4% DSS for 5 days (Figure 1A). After cessation of DSS

Discussion

Using a model of acute intestinal inflammation in which acquired immune responses can be abrogated, we here demonstrate that the pathophysiologic roles of TNFR1 and TNFR2 in innate immune pathways differ during acute intestinal inflammation. As observed in acquired immune responses,20 TNFR2-mediated activation of innate immune cells in the colonic LP contributes to the exacerbation of colitis. In contrast, TNFR1 expressing LP cells are required for epithelial regeneration following acute

References (34)

  • D.K. Podolsky

    Inflammatory bowel disease

    N Eng J Med

    (2002)
  • W. Strober et al.

    The immunology of mucosal models of inflammation

    Annu Rev Immunol

    (2002)
  • A. Mizoguchi et al.

    Immune networks in animal models of inflammatory bowel disease

    Inflamm Bowel Dis

    (2003)
  • E. Mizoguchi et al.

    Role of cytokines in the early stages of chronic colitis in TCRα-mutant mice

    Lab Invest

    (1997)
  • L. Mayer et al.

    Evidence for function of Ia molecules on gut epithelial cells in man

    J Exp Med

    (1987)
  • D. Wallach et al.

    Tumor necrosis factor receptor and Fas signaling mechanisms

    Annu Rev Immunol

    (1999)
  • G. Chen et al.

    TNF-R1 signaling: a beautiful pathway

    Science

    (2002)
  • Cited by (45)

    • TNF Receptor 1 Promotes Early-Life Immunity and Protects against Colitis in Mice

      2020, Cell Reports
      Citation Excerpt :

      However, the therapeutic effects of anti-TNF agents are short lived for many (Ben-Horin et al., 2014; Colombel et al., 2007; Ford et al., 2011; Schreiber et al., 2007). In mice, deletion of TNF or either of its transmembrane receptors, TNFR1 (p55) and TNFR2 (p75) (encoded by the Tnfrsf1a and Tnfrsf1b genes, respectively), has differing effects on disease susceptibility in established colitis models (Dubé et al., 2015; Ebach et al., 2005; Mizoguchi et al., 2008; Wang et al., 2012; Wang et al., 2013). Strikingly, loss of TNF-signaling-pathway members Tnf (Hale and Greer, 2012) or Tnfr2 (Punit et al., 2015) results in severe disease in the Il10−/− spontaneous colitis model (Kühn et al., 1993).

    • Interleukin 32, inflammation and cancer

      2017, Pharmacology and Therapeutics
      Citation Excerpt :

      Severity levels of DSS (dextran sodium sulfate)-induced colitis were lower in IL-32γ TG mice, compared with colitis in the wild type mice (Choi et al., 2010; Oh et al., 2011). Unexpectedly these results are similar in that DSS-induced colitis was acerbated in deficient mice of TNFα or TNFR1 compared to the wild type mice (Mizoguchi et al., 2008; Naito et al., 2003; Noti, Corazza, Mueller, Berger, & Brunner, 2010). These results suggest that IL-32 has a dual function in innate responses of the intestine, which constitutive expression of TNFα and IL-10 by IL-32 may regulate expression of inflammatory cytokine by NOD2 and protect the barrier from pathogens.

    • TNFR2 activates mlck-dependent tight junction dysregulation to cause apoptosis-mediated barrier loss and experimental colitis

      2013, Gastroenterology
      Citation Excerpt :

      This conclusion, which is consistent with in vitro data showing that TNFR2 signaling mediates TNF-induced long MLCK expression and tight junction regulation,6 might explain the association of TNFR2 polymorphisms with Crohn’s disease.35 Although a protective effect of TNFR2 knockout has recently been reported in DSS colitis,36 this result is controversial, as other studies have demonstrated no significant change or increased DSS sensitivity in TNFR2 knockout mice.37,38 One possible explanation for this disparity between studies is that differences in mouse strain used, local microbiome, or other factors that affect DSS colitis development highlighted or suppressed the well known role of TNFR2 in promoting epithelial migration and wound healing.8

    • Animal Models of Inflammatory Bowel Disease for Drug Discovery

      2013, Animal Models for the Study of Human Disease
    View all citing articles on Scopus

    Conflicts of interest: None of the authors have conflicts of interest to disclose.

    Supported by NIH grants DK64289, DK74454 (to E.M.), DK43351, DK60049 (to D.K.P.), and DK 64351 (to A.M.), and IBD Grants from Broad Medical Foundation (to E.M.).

    View full text