Paneth cells as a site of origin for intestinal inflammation

Timon E Adolph, Michal F Tomczak, Lukas Niederreiter, Hyun-Jeong Ko, Janne Böck, Eduardo Martinez-Naves, Jonathan N Glickman, Markus Tschurtschenthaler, John Hartwig, Shuhei Hosomi, Magdalena B Flak, Jennifer L Cusick, Kenji Kohno, Takao Iwawaki, Susanne Billmann-Born, Tim Raine, Richa Bharti, Ralph Lucius, Mi-Na Kweon, Stefan J Marciniak, Augustine Choi, Susan J Hagen, Stefan Schreiber, Philip Rosenstiel, Arthur Kaser, Richard S Blumberg, Timon E Adolph, Michal F Tomczak, Lukas Niederreiter, Hyun-Jeong Ko, Janne Böck, Eduardo Martinez-Naves, Jonathan N Glickman, Markus Tschurtschenthaler, John Hartwig, Shuhei Hosomi, Magdalena B Flak, Jennifer L Cusick, Kenji Kohno, Takao Iwawaki, Susanne Billmann-Born, Tim Raine, Richa Bharti, Ralph Lucius, Mi-Na Kweon, Stefan J Marciniak, Augustine Choi, Susan J Hagen, Stefan Schreiber, Philip Rosenstiel, Arthur Kaser, Richard S Blumberg

Abstract

The recognition of autophagy related 16-like 1 (ATG16L1) as a genetic risk factor has exposed the critical role of autophagy in Crohn's disease. Homozygosity for the highly prevalent ATG16L1 risk allele, or murine hypomorphic (HM) activity, causes Paneth cell dysfunction. As Atg16l1(HM) mice do not develop spontaneous intestinal inflammation, the mechanism(s) by which ATG16L1 contributes to disease remains obscure. Deletion of the unfolded protein response (UPR) transcription factor X-box binding protein-1 (Xbp1) in intestinal epithelial cells, the human orthologue of which harbours rare inflammatory bowel disease risk variants, results in endoplasmic reticulum (ER) stress, Paneth cell impairment and spontaneous enteritis. Unresolved ER stress is a common feature of inflammatory bowel disease epithelium, and several genetic risk factors of Crohn's disease affect Paneth cells. Here we show that impairment in either UPR (Xbp1(ΔIEC)) or autophagy function (Atg16l1(ΔIEC) or Atg7(ΔIEC)) in intestinal epithelial cells results in each other's compensatory engagement, and severe spontaneous Crohn's-disease-like transmural ileitis if both mechanisms are compromised. Xbp1(ΔIEC) mice show autophagosome formation in hypomorphic Paneth cells, which is linked to ER stress via protein kinase RNA-like endoplasmic reticulum kinase (PERK), elongation initiation factor 2α (eIF2α) and activating transcription factor 4 (ATF4). Ileitis is dependent on commensal microbiota and derives from increased intestinal epithelial cell death, inositol requiring enzyme 1α (IRE1α)-regulated NF-κB activation and tumour-necrosis factor signalling, which are synergistically increased when autophagy is deficient. ATG16L1 restrains IRE1α activity, and augmentation of autophagy in intestinal epithelial cells ameliorates ER stress-induced intestinal inflammation and eases NF-κB overactivation and intestinal epithelial cell death. ER stress, autophagy induction and spontaneous ileitis emerge from Paneth-cell-specific deletion of Xbp1. Genetically and environmentally controlled UPR function within Paneth cells may therefore set the threshold for the development of intestinal inflammation upon hypomorphic ATG16L1 function and implicate ileal Crohn's disease as a specific disorder of Paneth cells.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. PERK/eIF2α signaling induces autophagy in…
Figure 1. PERK/eIF2α signaling induces autophagy in Xbp1-deficient intestinal epithelial cells
a, b, Immunoblot for LC3 conversion in isolated primary IECs (a) (n=5/4) and for autophagy proteins in primary IEC scrapings (b) (n=3). c, Transmission electron microscopy (TEM) of crypts. Note autophagic vacuoles in various stages of evolution in Xbp1ΔIEC hypomorphic Paneth cells. d, e, Crypt showing GFP-LC3 punctae (d), quantified in (e) (n=10; unpaired Student’s t-test). Bar, 5 μm. f, g, Immunoblot of silenced MODE-K cells (f) and primary IEC scrapings (g) for the PERK/eIF2α branch (n=3). h, i, Promoter sequence qPCR for Map1lc3b (LC3b) (h) and Atg7 (i) after anti-ATF4 ChIP (unpaired Student’s t-test). j, GFP-LC3 punctae per crypt after treatment with tamoxifen for 3 days and vehicle or salubrinal (n=10; one-way ANOVA with post-hoc Bonferroni). (k) Enteritis histology score after salubrinal and tamoxifen co-treatment (n=12/14/13; median shown; Kruskal-Wallis with post-hoc Holm’s-corrected Mann-Whitney U). Results represent three (a, f, g) or two (c, e, h, i) independent experiments. *P < 0.05, ***P < 0.001.
Figure 2. Impairment of ER stress-induced compensatory…
Figure 2. Impairment of ER stress-induced compensatory autophagy results in severe transmural inflammation
a, TEM of crypts of Lieberkühn (n=2). Bar, 2 μm. b, Representative H&E stainings. Note transmural inflammation extending through muscularis propria (white arrow) into serosa (black arrow) in Atg7/Xbp1ΔIEC and Atg16l1/Xbp1ΔIEC mice scored in (c) and (e). Bar, 50 and 10 μm, respectively. c, Enteritis histology score (n=26/12/18/27; 10-18 weeks; median shown; Kruskal-Wallis with post-hoc Holm’s-corrected Mann-Whitney U). d, Crypt TEM in indicated genotypes (n=2). Bar, 2 μm. e, Enteritis histology score (n=11; 18 weeks; median shown; Kruskal-Wallis with post-hoc Holm’s-corrected Mann-Whitney U). f,Xbp1 mRNA splicing (Xbp1u, unspliced; Xbp1s, spliced) of crypts, densitometry in (g) (n=7; unpaired Student’s t-test). h, GRP78 (green) immunofluorescence, white arrows indicate GRP78+ crypts (DAPI, blue; n=5). Bar, 10 μm. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 3. Autophagy restrains IRE1α-mediated NFκB activation…
Figure 3. Autophagy restrains IRE1α-mediated NFκB activation in Xbp1-deficient epithelium
a,shCtrl or shXbp1 MODE-K cells were co-silenced for Atg16l1 (siAtg16l1) or with scrambled siRNA (siCtrl), and analyzed by flow cytometry for annexin V and propidium iodide (PI; one-way ANOVA with post-hoc Bonferroni). b, Immunoblot of primary IEC scrapings (n=3). c, Immunoblot of cytoplasmic extracts from shCtrl or shXbp1 MODE-K cells after TNF stimulation. d, TUNEL+ IECs per 100 crypts after BAY11-7082 or vehicle treatment (n=3/4/4; one-way ANOVA with post-hoc Holm’s-corrected unpaired Student’s t-test). e, Enteritis histology score of mice treated with BAY11-7082 or vehicle (n=10/10/9; median shown; Kruskal-Wallis with post-hoc Holm’s-corrected Mann-Whitney U). f, Immunoblot of IEC scrapings for (p-)IRE1α after IRE1α immunoprecipitation (IP). β-actin, loading control of whole lysates. g, Immunoblot of primary IEC scrapings (n=4). h, i, Enteritis histology score of indicated genotypes (h,n=15/16/14/15; i,n=5/10/12; median shown; Kruskal-Wallis with post-hoc Holm’s-corrected Mann-Whitney U). j, Enteritis histology score of specific pathogen free (SPF) and germ free (GF) housed mice (n=10/9/7/7; median shown; Kruskal-Wallis with post-hoc Holm’s-corrected Mann-Whitney U). (k) Representative images of p-IκBα immunoreactivity under conditions as in (j) (n=4). Bar, 20 μm. Results represent four (f), three (c) or two (a, b) independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 4. ER stress-induced enteritis originates from…
Figure 4. ER stress-induced enteritis originates from Paneth cells and is alleviated through autophagy induction
a, Immunoblot of primary IEC scrapings from mice treated with or without rapamycin for 14 consecutive days (n=3). b, Enteritis histology score for experiment as in (a) (n=4; median shown; Mann-Whitney U). c, Representative images of EYFP-Rosa26/D6-Cre+/− reporter mice and EYFP-Rosa26 (controls). Co-localization of Defa6 Cre-driven EYFP expression (yellow) with lysozyme expressing Paneth cells (red; n=3). DAPI, blue; bar, 50 μm. d, Immunoblots of crypt IECs from Xbp1ΔPC and Wt controls (n=2). e, f, Representative confocal images of lysozyme (green) expressing Paneth cells (e) with quantification of crypts with indicated number of lysozyme+ granulated dots in (f) (n=5; unpaired Student’s t-test). DAPI, blue; bar, 10 μm. g, Immunohistochemistry for p-eIF2α (n=3). Bar, 20 μm. h, Representative H&E images of Xbp1ΔPC and Wt mice scored in (i). Bar, 50 μm. (i) Enteritis scoring in Xbp1fl/fl (Wtfl), Defa6 Cre+ (WtCre) and Defa6 Cre+;Xbp1fl/fl (Xbp1ΔPC) mice (n=21/26/29; median shown; Kruskal-Wallis with post-hoc Holm’s-corrected Mann-Whitney U). Results represent two (b, d) independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001.

References

    1. Jostins L, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119–124.
    1. Cadwell K, et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature. 2008;456:259–263.
    1. Cadwell K, et al. Virus-plus-susceptibility gene interaction determines Crohn’s disease gene Atg16L1 phenotypes in intestine. Cell. 2010;141:1135–1145.
    1. Kaser A, et al. XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell. 2008;134:743–756.
    1. Treton X, et al. Altered endoplasmic reticulum stress affects translation in inactive colon tissue from patients with ulcerative colitis. Gastroenterology. 2011;141:1024–1035.
    1. Kobayashi KS, et al. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 2005;307:731–734.
    1. Zheng W, et al. Evaluation of AGR2 and AGR3 as candidate genes for inflammatory bowel disease. Genes Immun. 2006;7:11–18.
    1. Wehkamp J, et al. Reduced Paneth cell alpha-defensins in ileal Crohn’s disease. Proc Natl Acad Sci U S A. 2005;102:18129–18134.
    1. Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474:298–306.
    1. Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature. 2011;469:323–335.
    1. Sarkar S, Ravikumar B, Rubinsztein DC. Autophagic clearance of aggregate-prone proteins associated with neurodegeneration. Methods Enzymol. 2009;453:83–110.
    1. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell. 2004;15:1101–1111.
    1. Rouschop KM, et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J Clin Invest. 2010;120:127–141.
    1. Komatsu M, et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol. 2005;169:425–434.
    1. Boyce M, et al. A selective inhibitor of eIF2alpha dephosphorylation protects cells from ER stress. Science. 2005;307:935–939.
    1. Novoa I, et al. Stress-induced gene expression requires programmed recovery from translational repression. EMBO J. 2003;22:1180–1187.
    1. Hampe J, et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet. 2007;39:207–211.
    1. Cleynen I, et al. Genetic factors conferring an increased susceptibility to develop Crohn’s disease also influence disease phenotype: results from the IBDchip European Project. Gut. 2012
    1. Fujita N, et al. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell. 2008;19:2092–2100.
    1. Mizushima N, et al. Mouse Apg16L, a novel WD-repeat protein, targets to the autophagic isolation membrane with the Apg12-Apg5 conjugate. J Cell Sci. 2003;116:1679–1688.
    1. Fujita N, et al. Differential involvement of Atg16L1 in Crohn disease and canonical autophagy: analysis of the organization of the Atg16L1 complex in fibroblasts. J Biol Chem. 2009;284:32602–32609.
    1. Yang L, Li P, Fu S, Calay ES, Hotamisligil GS. Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Cell metabolism. 2010;11:467–478.
    1. Bertolotti A, et al. Increased sensitivity to dextran sodium sulfate colitis in IRE1beta-deficient mice. J Clin Invest. 2001;107:585–593.
    1. Cao SS, et al. The unfolded protein response and chemical chaperones reduce protein misfolding and colitis in mice. Gastroenterology. 2013;144:989–1000. e1006.
    1. Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annu Rev Immunol. 2010;28:573–621.
    1. Rogler G, et al. Nuclear factor kappaB is activated in macrophages and epithelial cells of inflamed intestinal mucosa. Gastroenterology. 1998;115:357–369.
    1. Kaneko M, Niinuma Y, Nomura Y. Activation signal of nuclear factor-kappa B in response to endoplasmic reticulum stress is transduced via IRE1 and tumor necrosis factor receptor-associated factor 2. Biol Pharm Bull. 2003;26:931–935.
    1. Vaishnava S, Behrendt CL, Ismail AS, Eckmann L, Hooper LV. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc Natl Acad Sci U S A. 2008;105:20858–20863.
    1. Kuballa P, Huett A, Rioux JD, Daly MJ, Xavier RJ. Impaired autophagy of an intracellular pathogen induced by a Crohn’s disease associated ATG16L1 variant. PLoS One. 2008;3:e3391.
    1. Deuring JJ, et al. Genomic ATG16L1 risk allele-restricted Paneth cell ER stress in quiescent Crohn’s disease. Gut. 2013 in press.
    1. Madison BB, et al. Cis elements of the villin gene control expression in restricted domains of the vertical (crypt) and horizontal (duodenum, cecum) axes of the intestine. J Biol Chem. 2002;277:33275–33283.
    1. Shimshek DR, et al. Codon-improved Cre recombinase (iCre) expression in the mouse. Genesis. 2002;32:19–26.
    1. Garabedian EM, Roberts LJ, McNevin MS, Gordon JI. Examining the role of Paneth cells in the small intestine by lineage ablation in transgenic mice. J Biol Chem. 1997;272:23729–23740.
    1. Bry L, et al. Paneth cell differentiation in the developing intestine of normal and transgenic mice. Proc Natl Acad Sci U S A. 1994;91:10335–10339.
    1. Srinivas S, et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev Biol. 2001;1:4.
    1. Iwawaki T, Akai R, Yamanaka S, Kohno K. Function of IRE1 alpha in the placenta is essential for placental development and embryonic viability. Proc Natl Acad Sci U S A. 2009;106:16657–16662.
    1. Baert L, et al. Detection of murine norovirus 1 by using plaque assay, transfection assay, and real-time reverse transcription-PCR before and after heat exposure. Appl Environ Microbiol. 2008;74:543–546.
    1. Pierce JW, et al. Novel inhibitors of cytokine-induced IkappaBalpha phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo. J Biol Chem. 1997;272:21096–21103.
    1. Satoh Y, Yamano M, Matsuda M, Ono K. Ultrastructure of Paneth cells in the intestine of various mammals. J Electron Microsc Tech. 1990;16:69–80.
    1. Vidal K, Grosjean I, evillard JP, Gespach C, Kaiserlian D. Immortalization of mouse intestinal epithelial cells by the SV40-large T gene. Phenotypic and immune characterization of the MODE-K cell line. J Immunol Methods. 1993;166:63–73.
    1. Hempel SL, Buettner GR, O’Malley YQ, Wessels DA, Flaherty DM. Dihydrofluorescein diacetate is superior for detecting intracellular oxidants: comparison with 2’,7’-dichlorodihydrofluorescein diacetate, 5(and 6)-carboxy-2’,7’-dichlorodihydrofluorescein diacetate, and dihydrorhodamine 123. Free Radic Biol Med. 1999;27:146–159.
    1. Sato T, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–265.
    1. Wei Y, Sinha S, Levine B. Dual role of JNK1-mediated phosphorylation of Bcl-2 in autophagy and apoptosis regulation. Autophagy. 2008;4:949–951.
    1. Carchman EH, Rao J, Loughran PA, Rosengart MR, Zuckerbraun BS. Heme oxygenase-1-mediated autophagy protects against hepatocyte cell death and hepatic injury from infection/sepsis in mice. Hepatology. 2011;53:2053–2062.
    1. Hu P, Han Z, Couvillon AD, Kaufman RJ, Exton JH. Autocrine tumor necrosis factor alpha links endoplasmic reticulum stress to the membrane death receptor pathway through IRE1alpha-mediated NF-kappaB activation and down-regulation of TRAF2 expression. Mol Cell Biol. 2006;26:3071–3084.
    1. Urano F, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 2000;287:664–666.
    1. Barrett JC, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn’s disease. Nat Genet. 2008;40:955–962.
    1. Tashiro E, et al. Trierixin, a novel Inhibitor of ER stress-induced XBP1 activation from Streptomyces sp. 1. Taxonomy, fermentation, isolation and biological activities. J Antibiot (Tokyo) 2007;60:547–553.
    1. McKinney EF, et al. A CD8+ T cell transcription signature predicts prognosis in autoimmune disease. Nat Med. 2010;16:586–591. 581p following 591.
    1. Rioux JD, et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet. 2007;39:596–604.
    1. Parkes M, et al. Sequence variants in the autophagy gene IRGM and multiple other replicating loci contribute to Crohn’s disease susceptibility. Nat Genet. 2007;39:830–832.

Source: PubMed

3
구독하다