174
Views
3
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

The Deletion of IL-17A Enhances Helicobacter hepaticus Colonization and Triggers Colitis

, , , , , & ORCID Icon show all
Pages 2761-2773 | Published online: 29 Apr 2022

References

  • Ananthakrishnan AN, Bernstein CN, Iliopoulos D, et al. Environmental triggers in IBD: a review of progress and evidence. Nat Rev Gastroenterol Hepatol. 2018;15(1):39–49. doi:10.1038/nrgastro.2017.136
  • de Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nat Rev Gastroenterol Hepatol. 2016;13(1):13–27. doi:10.1038/nrgastro.2015.186
  • Lavelle A, Sokol H. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17(4):223–237. doi:10.1038/s41575-019-0258-z
  • Fox JG, Ge Z, Whary MT, Erdman SE, Horwitz BH. Helicobacter hepaticus infection in mice: models for understanding lower bowel inflammation and cancer. Mucosal Immunol. 2011;4(1):22–30. doi:10.1038/mi.2010.61
  • Fujino S, Andoh A, Bamba S, et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut. 2003;52(1):65–70. doi:10.1136/gut.52.1.65
  • Cao S, Zhu C, Feng J, et al. Helicobacter hepaticus infection induces chronic hepatitis and fibrosis in male BALB/c mice via the activation of NF-κB, Stat3, and MAPK signaling pathways. Helicobacter. 2020;25(2):e12677. doi:10.1111/hel.12677
  • Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–498. doi:10.1016/j.cell.2009.09.033
  • Rouvier E, Luciani MF, Mattéi MG, Denizot F, Golstein P. CTLA-8, cloned from an activated T cell, bearing AU-rich messenger RNA instability sequences, and homologous to a herpesvirus saimiri gene. J Immunol. 1993;150(12):5445–5456.
  • Amatya N, Garg AV, Gaffen SL. IL-17 Signaling: the Yin and the Yang. Trends Immunol. 2017;38(5):310–322. doi:10.1016/j.it.2017.01.006
  • Onishi RM, Gaffen SL. Interleukin-17 and its target genes: mechanisms of interleukin-17 function in disease. Immunology. 2010;129(3):311–321. doi:10.1111/j.1365-2567.2009.03240.x
  • Kim K, Kim G, Kim J-Y, Yun HJ, Lim S-C, Choi HS. Interleukin-22 promotes epithelial cell transformation and breast tumorigenesis via MAP3K8 activation. Carcinogenesis. 2014;35(6):1352–1361. doi:10.1093/carcin/bgu044
  • Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional specialization of interleukin-17 family members. Immunity. 2011;34(2):149–162. doi:10.1016/j.immuni.2011.02.012
  • Kinugasa T, Sakaguchi T, Gu X, Reinecker HC. Claudins regulate the intestinal barrier in response to immune mediators. Gastroenterology. 2000;118(6):1001–1011. doi:10.1016/S0016-5085(00)70351-9
  • Qin H, Tang G, Yi P, et al. Diagnosis of Genus Helicobacter through a hemi-nested PCR assay of 16S rRNA. Saudi Pharm J. 2016;24(3):265–272. doi:10.1016/j.jsps.2016.04.015
  • Xu M, Pokrovskii M, Ding Y, et al. c-MAF-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont. Nature. 2018;554(7692):373–377. doi:10.1038/nature25500
  • Ge Z, Feng Y, Whary MT, et al. Cytolethal distending toxin is essential for Helicobacter hepaticus colonization in outbred Swiss Webster mice. Infect Immun. 2005;73(6):3559–3567. doi:10.1128/IAI.73.6.3559-3567.2005
  • Kitajima S, Takuma S, Morimoto M. Changes in colonic mucosal permeability in mouse colitis induced with dextran sulfate sodium. Exp Anim. 1999;48(3):137–143. doi:10.1538/expanim.48.137
  • Cao S, Zhu L, Zhu C, et al. Helicobacter hepaticus infection-induced IL-33 promotes hepatic inflammation and fibrosis through ST2 signaling pathways in BALB/c mice. Biochem Biophys Res Commun. 2020;525(3):654–661. doi:10.1016/j.bbrc.2020.02.139
  • Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–408. doi:10.1006/meth.2001.1262
  • Buonocore S, Ahern PP, Uhlig HH, et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature. 2010;464(7293):1371–1375. doi:10.1038/nature08949
  • Friedrich V, Forné I, Matzek D, et al. Helicobacter hepaticus is required for immune targeting of bacterial heat shock protein 60 and fatal colitis in mice. Gut Microbes. 2021;13(1):1–20. doi:10.1080/19490976.2021.1882928
  • Han X, Huang T, Han J. Cytokines derived from innate lymphoid cells assist Helicobacter hepaticus to aggravate hepatocellular tumorigenesis in viral transgenic mice. Gut Pathog. 2019;11:23. doi:10.1186/s13099-019-0302-0
  • Morrison PJ, Ballantyne SJ, Macdonald SJ, et al. Differential requirements for IL-17A and IL-22 in cecal versus colonic inflammation induced by Helicobacter hepaticus. Am J Pathol. 2015;185(12):3290–3303. doi:10.1016/j.ajpath.2015.08.015
  • Bevins CL, Salzman NH. Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nat Rev Microbiol. 2011;9(5):356–368. doi:10.1038/nrmicro2546
  • Yu S, Balasubramanian I, Laubitz D, et al. Paneth cell-derived lysozyme defines the composition of mucolytic microbiota and the inflammatory tone of the intestine. Immunity. 2020;53(2):398–416 e398. doi:10.1016/j.immuni.2020.07.010
  • Gordon S, Todd J, Cohn ZA. In vitro synthesis and secretion of lysozyme by mononuclear phagocytes. J Exp Med. 1974;139(5):1228–1248. doi:10.1084/jem.139.5.1228
  • Ralph P, Moore MA, Nilsson K. Lysozyme synthesis by established human and murine histiocytic lymphoma cell lines. J Exp Med. 1976;143(6):1528–1533. doi:10.1084/jem.143.6.1528
  • Gao X, Guo M, Zhang Z, Shen P, Yang Z, Zhang N. Baicalin promotes the bacteriostatic activity of lysozyme on S. aureus in mammary glands and neutrophilic granulocytes in mice. Oncotarget. 2017;8(12):19894–19901. doi:10.18632/oncotarget.15193
  • Danne C, Ryzhakov G, Martínez-López M, et al. A large polysaccharide produced by Helicobacter hepaticus induces an anti-inflammatory gene signature in macrophages. Cell Host Microbe. 2017;22(6):733–745.e5. doi:10.1016/j.chom.2017.11.002
  • Zhu L, Zhu C, Cao S, Zhang Q. Helicobacter hepaticus induce colitis in male IL-10(-/-) mice dependent by cytolethal distending toxin B and via the activation of jak/stat signaling pathway. Front Cell Infect Microbiol. 2021;11:616218. doi:10.3389/fcimb.2021.616218
  • Fauny M, Moulin D, D’Amico F, et al. Paradoxical gastrointestinal effects of interleukin-17 blockers. Ann Rheum Dis. 2020;79(9):1132–1138. doi:10.1136/annrheumdis-2020-217927
  • Li J, Huang L, Zhao H, Yan Y, Lu J. The role of interleukins in colorectal cancer. Int J Biol Sci. 2020;16(13):2323–2339. doi:10.7150/ijbs.46651
  • Marks BR, Craft J. Barrier immunity and IL-17. Semin Immunol. 2009;21(3):164–171. doi:10.1016/j.smim.2009.03.001
  • Brevi A, Cogrossi LL, Grazia G, et al. Much more than IL-17A: cytokines of the IL-17 family between microbiota and cancer. Front Immunol. 2020;11:565470. doi:10.3389/fimmu.2020.565470
  • Benakis C, Brea D, Caballero S, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal gammadelta T cells. Nat Med. 2016;22(5):516–523. doi:10.1038/nm.4068
  • Stockinger B, Omenetti S. The dichotomous nature of T helper 17 cells. Nat Rev Immunol. 2017;17(9):535–544. doi:10.1038/nri.2017.50
  • Liang SC, Tan XY, Luxenberg DP, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. 2006;203(10):2271–2279. doi:10.1084/jem.20061308
  • Kuwabara T, Ishikawa F, Kondo M, Kakiuchi T. The role of IL-17 and related cytokines in inflammatory autoimmune diseases. Mediators Inflamm. 2017;2017:3908061. doi:10.1155/2017/3908061
  • Bergamo A, Gerdol M, Pallavicini A, et al. Lysozyme-induced transcriptional regulation of TNF-alpha pathway genes in cells of the monocyte lineage. Int J Mol Sci. 2019;20:21. doi:10.3390/ijms20215502
  • Warfel AH, Zucker-Franklin D. Down-regulation of macrophage lysozyme by lipopolysaccharide and interferon. J Immunol. 1986;137(2):651–655.
  • Schoeniger A, Adolph S, Fuhrmann H, Schumann J. The impact of membrane lipid composition on macrophage activation in the immune defense against Rhodococcus equi and Pseudomonas aeruginosa. Int J Mol Sci. 2011;12(11):7510–7528. doi:10.3390/ijms12117510
  • Singh SB, Lin HC. Autophagy counters LPS-mediated suppression of lysozyme. Innate Immun. 2017;23(6):537–545. doi:10.1177/1753425917721630
  • He Z, Gharaibeh RZ, Newsome RC, et al. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut. 2019;68(2):289–300. doi:10.1136/gutjnl-2018-317200
  • Ge Z, Feng Y, Ge L, Parry N, Muthupalani S, Fox JG. Helicobacter hepaticus cytolethal distending toxin promotes intestinal carcinogenesis in 129 Rag2 -deficient mice. Cell Microbiol. 2017;19(7):e12728. doi:10.1111/cmi.12728
  • Dudakov JA, Hanash AM, van den Brink MR. Interleukin-22: immunobiology and pathology. Annu Rev Immunol. 2015;33:747–785. doi:10.1146/annurev-immunol-032414-112123
  • Hasnain SZ, Tauro S, Das I, et al. IL-10 promotes production of intestinal mucus by suppressing protein misfolding and endoplasmic reticulum stress in goblet cells. Gastroenterology. 2013;144(2):357–368 e359. doi:10.1053/j.gastro.2012.10.043
  • Nishida A, Lau CW, Zhang M, et al. The membrane-bound mucin Muc1 regulates T helper 17-cell responses and colitis in mice. Gastroenterology. 2012;142(4):865–874 e862. doi:10.1053/j.gastro.2011.12.036
  • Lagow EL, Carson DD. Synergistic stimulation of MUC1 expression in normal breast epithelia and breast cancer cells by interferon-gamma and tumor necrosis factor-alpha. J Cell Biochem. 2002;86(4):759–772. doi:10.1002/jcb.10261
  • Lindemans CA, Calafiore M, Mertelsmann AM, et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature. 2015;528(7583):560–564. doi:10.1038/nature16460