28
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      A Novel Role for Necroptosis in the Pathogenesis of Necrotizing Enterocolitis

      research-article
      1 , 2 , 2 , 2 , 2 , 2 , 2 , 3 , 4 , 4 , 2 , 2 , 2 , 2 , 2 , 2 , ∗∗ , 2 , 3 ,
      Cellular and Molecular Gastroenterology and Hepatology
      Elsevier
      Pediatrics, Premature, Organoid Model, diff, differentiated, HMGB1, high mobility group box 1, IEC, intestinal epithelial cell, IHC, immunohistochemistry, IL, interleukin, LPS, lipopolysaccharide, Mlkl, mixed lineage kinase domain-like protein, NEC, necrotizing enterocolitis, p, postnatal day, PCNA, proliferating cell nuclear antigen, pRIPK, phosphorylated receptor-interacting serine/threonine-protein kinase, qRT-PCR, real-time quantitative reverse-transcription polymerase chain reaction, Ripk, receptor-interacting serine/threonine-protein kinase, ROI, region of interest, TBST, Tris-buffered saline with 0.1% Tween-20, TLR4, Toll-like receptor 4, TNF, tumor necrosis factor, undiff, undifferentiated, 2’FL, 2'-fucosyllactose, 3NT, 3-nitrotyrosine

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background & Aims

          Necrotizing enterocolitis (NEC) is a devastating disease of premature infants characterized by Toll-like receptor 4 (TLR4)-dependent intestinal inflammation and enterocyte death. Given that necroptosis is a proinflammatory cell death process that is linked to bacterial signaling, we investigated its potential role in NEC, and the mechanisms involved.

          Methods

          Human and mouse NEC intestine were analyzed for necroptosis gene expression (ie, RIPK1, RIPK3, and MLKL), and protein activation (phosphorylated RIPK3). To evaluate a potential role for necroptosis in NEC, the effects of genetic (ie, Ripk3 knockout or Mlkl knockout) or pharmacologic (ie, Nec1s) inhibition of intestinal inflammation were assessed in a mouse NEC model, and a possible upstream role of TLR4 was assessed in Tlr4-deficient mice. The NEC-protective effects of human breast milk and its constituent milk oligosaccharides on necroptosis were assessed in a NEC-in-a-dish model, in which mouse intestinal organoids were cultured as either undifferentiated or differentiated epithelium in the presence of NEC bacteria and hypoxia.

          Results

          Necroptosis was activated in the intestines of human and mouse NEC in a TLR4-dependent manner, and was up-regulated specifically in differentiated epithelium of the immature ileum. Inhibition of necroptosis genetically and pharmacologically reduced intestinal–epithelial cell death and mucosal inflammation in experimental NEC, and ex vivo in the NEC-in-a-dish system. Strikingly, the addition of human breast milk, or the human milk oligosaccharide 2 fucosyllactose in the ex vivo system, reduced necroptosis and inflammation.

          Conclusions

          Necroptosis is activated in the intestinal epithelium upon TLR4 signaling and is required for NEC development, and explains in part the protective effects of breast milk.

          Graphical abstract

          Related collections

          Most cited references46

          • Record: found
          • Abstract: found
          • Article: not found

          Caspase-8 regulates TNF-alpha induced epithelial necroptosis and terminal ileitis

          Dysfunction of the intestinal epithelium is believed to result in excessive translocation of commensal bacteria into the bowel wall that drives chronic mucosal inflammation in Crohn's disease; an incurable inflammatory bowel disease in humans characterized by inflammation of the terminal ileum 1 . Beside the physical barrier established by the tight contact of cells, specialized epithelial cells such as Paneth cells and goblet cells provide innate immune defence functions by secreting mucus and antimicrobial peptides which hamper access and survival of bacteria adjacent to the epithelium 2 . Epithelial cell death is a hallmark of intestinal inflammation and has been discussed as a pathogenic mechanism driving Crohn's disease (CD) in humans 3 . However, the regulation of epithelial cell death and its role in intestinal homeostasis remains poorly understood. Here we demonstrate a critical role for caspase-8 in regulating necroptosis of intestinal epithelial cells (IEC) and terminal ileitis. Mice with a conditional deletion of caspase-8 in the intestinal epithelium (Casp8 ΔIEC) spontaneously developed inflammatory lesions in the terminal ileum and were highly susceptible to colitis. Casp8 ΔIEC mice lacked Paneth cells and showed reduced numbers of goblet cells suggesting dysregulated anti-microbial immune cell functions of the intestinal epithelium. Casp8 ΔIEC mice showed increased cell death in the Paneth cell area of small intestinal crypts. Epithelial cell death was induced by tumor necrosis factor (TNF) -α, was associated with increased expression of receptor-interacting protein 3 (RIP3) and could be inhibited upon blockade of necroptosis. Finally, we identified high levels of RIP3 in human Paneth cells and increased necroptosis in the terminal ileum of patients with Crohn's disease, suggesting a potential role of necroptosis in the pathogenesis of this disease. Taken together, our data demonstrate a critical function of caspase-8 in regulating intestinal homeostasis and in protecting IEC from TNF-α induced necroptotic cell death.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic cell death.

            Necroptosis is considered to be complementary to the classical caspase-dependent programmed cell death pathway, apoptosis. The pseudokinase Mixed Lineage Kinase Domain-Like (MLKL) is an essential effector protein in the necroptotic cell death pathway downstream of the protein kinase Receptor Interacting Protein Kinase-3 (RIPK3). How MLKL causes cell death is unclear, however RIPK3-mediated phosphorylation of the activation loop in MLKL trips a molecular switch to induce necroptotic cell death. Here, we show that the MLKL pseudokinase domain acts as a latch to restrain the N-terminal four-helix bundle (4HB) domain and that unleashing this domain results in formation of a high-molecular-weight, membrane-localized complex and cell death. Using alanine-scanning mutagenesis, we identified two clusters of residues on opposing faces of the 4HB domain that were required for the 4HB domain to kill cells. The integrity of one cluster was essential for membrane localization, whereas MLKL mutations in the other cluster did not prevent membrane translocation but prevented killing; this demonstrates that membrane localization is necessary, but insufficient, to induce cell death. Finally, we identified a small molecule that binds the nucleotide binding site within the MLKL pseudokinase domain and retards MLKL translocation to membranes, thereby preventing necroptosis. This inhibitor provides a novel tool to investigate necroptosis and demonstrates the feasibility of using small molecules to target the nucleotide binding site of pseudokinases to modulate signal transduction.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Kinase RIP3 Is Dispensable for Normal NF- Bs, Signaling by the B-Cell and T-Cell Receptors, Tumor Necrosis Factor Receptor 1, and Toll-Like Receptors 2 and 4

              Molecular and Cellular Biology, 24(4), 1464-1469
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Mol Gastroenterol Hepatol
                Cell Mol Gastroenterol Hepatol
                Cellular and Molecular Gastroenterology and Hepatology
                Elsevier
                2352-345X
                2020
                19 November 2019
                : 9
                : 3
                : 403-423
                Affiliations
                [1 ]Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
                [2 ]Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
                [3 ]McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
                [4 ]Abbott Nutrition, Columbus, Ohio
                Author notes
                [] Correspondence Address correspondence to: David Hackam, MD, Department of Surgery, Johns Hopkins University, The Johns Hopkins Children’s Center, Room 7323, 1800 Orleans Street, Baltimore, Maryland 21287. fax: (410) 502-5314. Dhackam1@ 123456jhmi.edu
                [∗∗ ]Chhinder Sodhi, PhD, Department of Surgery, Johns Hopkins University, Miller Research Building, 733 North Broadway, Room 470, Baltimore, Maryland 21205. fax: (410) 502-5314. csodhi@ 123456jhmi.edu
                Article
                S2352-345X(19)30157-2
                10.1016/j.jcmgh.2019.11.002
                7015998
                31756560
                8645efd8-c2bd-4074-b43f-c3aa4256ee87
                © 2020 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 21 June 2019
                : 12 November 2019
                Categories
                Original Research

                pediatrics,premature,organoid model,diff, differentiated,hmgb1, high mobility group box 1,iec, intestinal epithelial cell,ihc, immunohistochemistry,il, interleukin,lps, lipopolysaccharide,mlkl, mixed lineage kinase domain-like protein,nec, necrotizing enterocolitis,p, postnatal day,pcna, proliferating cell nuclear antigen,pripk, phosphorylated receptor-interacting serine/threonine-protein kinase,qrt-pcr, real-time quantitative reverse-transcription polymerase chain reaction,ripk, receptor-interacting serine/threonine-protein kinase,roi, region of interest,tbst, tris-buffered saline with 0.1% tween-20,tlr4, toll-like receptor 4,tnf, tumor necrosis factor,undiff, undifferentiated,2’fl, 2'-fucosyllactose,3nt, 3-nitrotyrosine

                Comments

                Comment on this article