Mmp17-deficient mice exhibit heightened goblet cell effector expression in the colon and increased resistance to chronic Trichuris muris infection

Intestinal epithelial homeostasis is maintained by intrinsic and extrinsic signals. The extrinsic signals include those provided by mesenchymal cell populations that surround intestinal crypts and is further facilitated by the extracellular matrix (ECM), which is modulated by proteases such as matrix metalloproteinases (MMPs). Extrinsic signals ensure an appropriate balance between intestinal epithelial proliferation and differentiation. This study explores the role of MMP17, which is expressed by mesenchymal cells, in intestinal homeostasis and during immunity to infection. Mice lacking MMP17 expressed high levels of goblet-cell associated genes, such as CLCA1 and RELM-β, which are normally associated with immune responses to infection. Nevertheless, Mmp17 KO mice did not have altered resistance during a bacterial Citrobacter rodentium infection. However, when challenged with a low dose of the helminth Trichuris muris, Mmp17 KO mice had increased resistance, without a clear role for an altered immune response during infection. Mechanistically, we did not find changes in traditional modulators of goblet cell effectors such as the NOTCH pathway or specific cytokines. Instead, we found elevated BMP signaling in Mmp17 KO mouse large intestinal crypts that we propose to alter the goblet cell maturation state. Together, our data suggests that MMP17 extrinsically alters the goblet cell maturation state via a BMP signaling axis, which is sufficient to alter clearance in a helminth infection model.

[1]  M. Johansson,et al.  The role of goblet cells and mucus in intestinal homeostasis , 2022, Nature Reviews Gastroenterology & Hepatology.

[2]  Mst. Farzana Sultana,et al.  Identification of Crucial Amino Acid Residues for Antimicrobial Activity of Angiogenin 4 and Its Modulation of Gut Microbiota in Mice , 2022, Frontiers in Microbiology.

[3]  Alberto Díez-Sánchez,et al.  BMP-signaling in the intestinal epithelium drives a critical feedback loop to restrain IL-13-driven tuft cell hyperplasia , 2022, Science Immunology.

[4]  A. van Oudenaarden,et al.  BMP gradient along the intestinal villus axis controls zonated enterocyte and goblet cell states. , 2022, Cell reports.

[5]  P. Katajisto,et al.  Smooth muscle-specific MMP17 (MT4-MMP) regulates the intestinal stem cell niche and regeneration after damage , 2021, Nature Communications.

[6]  Zheng Kuang,et al.  Small proline-rich protein 2A is a gut bactericidal protein deployed during helminth infection , 2021, Science.

[7]  Yun Han Kwon,et al.  Trichuris muris Model: Role in Understanding Intestinal Immune Response, Inflammation and Host Defense , 2021, Pathogens.

[8]  L. Boon,et al.  Trichuris muris infection drives cell-intrinsic IL4R alpha independent colonic RELMα+ macrophages , 2021, PLoS pathogens.

[9]  M. Bäckström,et al.  The IgGFc-binding protein FCGBP is secreted with all GDPH sequences cleaved but maintained by interfragment disulfide bonds , 2021, The Journal of biological chemistry.

[10]  A. Vetuschi,et al.  The Charming World of the Extracellular Matrix: A Dynamic and Protective Network of the Intestinal Wall , 2021, Frontiers in Medicine.

[11]  Y. Kluger,et al.  Paracrine orchestration of intestinal tumorigenesis by a mesenchymal niche , 2020, Nature.

[12]  Guocheng Yuan,et al.  Distinct Mesenchymal Cell Populations Generate the Essential Intestinal BMP Signaling Gradient. , 2020, Cell stem cell.

[13]  Cong-Lin Liu,et al.  Calcium-activated chloride channel regulator 1 (CLCA1): More than a regulator of chloride transport and mucus production☆ , 2019, The World Allergy Organization journal.

[14]  L. Arike,et al.  Calcium-activated chloride channel regulator 1 (CLCA1) forms non-covalent oligomers in colonic mucus and has mucin 2–processing properties , 2019, The Journal of Biological Chemistry.

[15]  M. Barrachina,et al.  Macrophages as an Emerging Source of Wnt Ligands: Relevance in Mucosal Integrity , 2019, Front. Immunol..

[16]  A. Gruber,et al.  Calcium-activated Chloride Channel Regulator 1 (CLCA1) Controls Mucus Expansion in Colon by Proteolytic Activity , 2018, EBioMedicine.

[17]  K. Basler,et al.  GLI1-expressing mesenchymal cells form the essential Wnt-secreting niche for colon stem cells , 2018, Nature.

[18]  K. Sigmundsson,et al.  PDGFRα+ pericryptal stromal cells are the critical source of Wnts and RSPO3 for murine intestinal stem cells in vivo , 2018, Proceedings of the National Academy of Sciences.

[19]  R. Grencis,et al.  A sticky end for gastrointestinal helminths; the role of the mucus barrier , 2018, Parasite immunology.

[20]  Vanessa Núñez,et al.  MT4-MMP deficiency increases patrolling monocyte recruitment to early lesions and accelerates atherosclerosis , 2018, Nature Communications.

[21]  S. Itzkovitz,et al.  Subepithelial telocytes are an important source of Wnts that supports intestinal crypts , 2018, Nature.

[22]  L. Hooper,et al.  Resistin-like molecule β is a bactericidal protein that promotes spatial segregation of the microbiota and the colonic epithelium , 2017, Proceedings of the National Academy of Sciences.

[23]  H. Miyoshi,et al.  Prostaglandin E2 promotes intestinal repair through an adaptive cellular response of the epithelium , 2017, The EMBO journal.

[24]  B. Dewals,et al.  RELMs in the Realm of Helminths. , 2016, Trends in parasitology.

[25]  K. Jakubowska,et al.  Expressions of Matrix Metalloproteinases (MMP-2, MMP-7, and MMP-9) and Their Inhibitors (TIMP-1, TIMP-2) in Inflammatory Bowel Diseases , 2016, Gastroenterology research and practice.

[26]  J. Redondo,et al.  Deficiency of MMP17/MT4-MMP proteolytic activity predisposes to aortic aneurysm in mice. , 2015, Circulation research.

[27]  Z. Werb,et al.  Matrix metalloproteinases in stem cell regulation and cancer , 2015, Matrix biology : journal of the International Society for Matrix Biology.

[28]  Hans Clevers,et al.  Plasticity within stem cell hierarchies in mammalian epithelia. , 2015, Trends in cell biology.

[29]  B. Finlay,et al.  Citrobacter rodentium: infection, inflammation and the microbiota , 2014, Nature Reviews Microbiology.

[30]  David Artis,et al.  Intestinal epithelial cells: regulators of barrier function and immune homeostasis , 2014, Nature Reviews Immunology.

[31]  M. Johansson,et al.  Studies of mucus in mouse stomach, small intestine, and colon. I. Gastrointestinal mucus layers have different properties depending on location as well as over the Peyer's patches. , 2013, American journal of physiology. Gastrointestinal and liver physiology.

[32]  M. Johansson,et al.  Studies of mucus in mouse stomach, small intestine, and colon. II. Gastrointestinal mucus proteome reveals Muc2 and Muc5ac accompanied by a set of core proteins. , 2013, American journal of physiology. Gastrointestinal and liver physiology.

[33]  R. Grencis,et al.  Trichuris muris: a model of gastrointestinal parasite infection , 2012, Seminars in Immunopathology.

[34]  Ruth Forman,et al.  The Goblet Cell Is the Cellular Source of the Anti-Microbial Angiogenin 4 in the Large Intestine Post Trichuris muris Infection , 2012, PloS one.

[35]  T. Wynn,et al.  Muc5ac: a critical component mediating the rejection of enteric nematodes , 2011, The Journal of experimental medicine.

[36]  R. Grencis,et al.  Changes in the mucosal barrier during acute and chronic Trichuris muris infection , 2011, Parasite immunology.

[37]  B. Finlay,et al.  Muc2 Protects against Lethal Infectious Colitis by Disassociating Pathogenic and Commensal Bacteria from the Colonic Mucosa , 2010, PLoS pathogens.

[38]  F. Finkelman,et al.  Intestinal epithelial cell secretion of RELM-β protects against gastrointestinal worm infection , 2009, The Journal of experimental medicine.

[39]  K. Else,et al.  Expulsion of Trichuris muris is associated with increased expression of angiogenin 4 in the gut and increased acidity of mucins within the goblet cell , 2009, BMC Genomics.

[40]  F. Shanahan,et al.  Involvement of T helper type 17 and regulatory T cell activity in Citrobacter rodentium invasion and inflammatory damage , 2009, Clinical and experimental immunology.

[41]  A. Murphy,et al.  Goblet Cell-Derived Resistin-Like Molecule β Augments CD4+ T Cell Production of IFN-γ and Infection-Induced Intestinal Inflammation1 , 2008, The Journal of Immunology.

[42]  H. Clevers,et al.  Identification of stem cells in small intestine and colon by marker gene Lgr5 , 2007, Nature.

[43]  N. Yoshida,et al.  Establishment of an MT4‐MMP‐deficient mouse strain representing an efficient tracking system for MT4‐MMP/MMP‐17 expression in vivo using β‐galactosidase , 2007, Genes to cells : devoted to molecular & cellular mechanisms.

[44]  Andrew J. Ewald,et al.  Matrix metalloproteinases and the regulation of tissue remodelling , 2007, Nature Reviews Molecular Cell Biology.

[45]  D. Artis New weapons in the war on worms: identification of putative mechanisms of immune-mediated expulsion of gastrointestinal nematodes. , 2006, International journal for parasitology.

[46]  G. Dougan,et al.  Citrobacter rodentium of mice and man , 2005, Cellular microbiology.

[47]  N. Paton,et al.  Identification of Novel Genes in Intestinal Tissue That Are Regulated after Infection with an Intestinal Nematode Parasite , 2005, Infection and Immunity.

[48]  M. Lazar,et al.  RELMbeta/FIZZ2 is a goblet cell-specific immune-effector molecule in the gastrointestinal tract. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[49]  K. Else,et al.  Low‐level infection with Trichuris muris significantly affects the polarization of the CD4 response , 1994, European journal of immunology.

[50]  E. Ruitenberg,et al.  The ‘Swiss roll’: a simple technique for histological studies of the rodent intestine , 1981, Laboratory animals.