Cargando…

Nonmicrobial Activation of TLRs Controls Intestinal Growth, Wound Repair, and Radioprotection

TLRs, key components of the innate immune system, recognize microbial molecules. However, TLRs also recognize some nonmicrobial molecules. In particular, TLR2 and TLR4 recognize hyaluronic acid, a glycosaminoglycan in the extracellular matrix. In neonatal mice endogenous hyaluronic acid binding to T...

Descripción completa

Detalles Bibliográficos
Autores principales: Stenson, William F., Ciorba, Matthew A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7859088/
https://www.ncbi.nlm.nih.gov/pubmed/33552081
http://dx.doi.org/10.3389/fimmu.2020.617510
_version_ 1783646658839445504
author Stenson, William F.
Ciorba, Matthew A.
author_facet Stenson, William F.
Ciorba, Matthew A.
author_sort Stenson, William F.
collection PubMed
description TLRs, key components of the innate immune system, recognize microbial molecules. However, TLRs also recognize some nonmicrobial molecules. In particular, TLR2 and TLR4 recognize hyaluronic acid, a glycosaminoglycan in the extracellular matrix. In neonatal mice endogenous hyaluronic acid binding to TLR4 drives normal intestinal growth. Hyaluronic acid binding to TLR4 in pericryptal macrophages results in cyclooxygenase2- dependent PGE(2) production, which transactivates EGFR in LGR5+ crypt epithelial stem cells leading to increased proliferation. The expanded population of LGR5+ stem cells leads to crypt fission and lengthening of the intestine and colon. Blocking this pathway at any point (TLR4 activation, PGE(2) production, EGFR transactivation) results in diminished intestinal and colonic growth. A similar pathway leads to epithelial proliferation in wound repair. The repair phase of dextran sodium sulfate colitis is marked by increased epithelial proliferation. In this model, TLR2 and TLR4 in pericryptal macrophages are activated by microbial products or by host hyaluronic acid, resulting in production of CXCL12, a chemokine. CXCL12 induces the migration of cyclooxygenase2-expressing mesenchymal stem cells from the lamina propria of the upper colonic crypts to a site adjacent to LGR5+ epithelial stem cells. PGE(2) released by these mesenchymal stem cells transactivates EGFR in LGR5+ epithelial stem cells leading to increased proliferation. Several TLR2 and TLR4 agonists, including hyaluronic acid, are radioprotective in the intestine through the inhibition of radiation-induced apoptosis in LGR5+ epithelial stem cells. Administration of exogenous TLR2 or TLR4 agonists activates TLR2/TLR4 on pericryptal macrophages inducing CXCL12 production with migration of cyclooxygenase2-expressing mesenchymal stem cells from the lamina propria of the villi to a site adjacent to LGR5+ epithelial stem cells. PGE(2) produced by these mesenchymal stem cells, blocks radiation-induced apoptosis in LGR5+ epithelial stem cells by an EGFR mediated pathway.
format Online
Article
Text
id pubmed-7859088
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-78590882021-02-05 Nonmicrobial Activation of TLRs Controls Intestinal Growth, Wound Repair, and Radioprotection Stenson, William F. Ciorba, Matthew A. Front Immunol Immunology TLRs, key components of the innate immune system, recognize microbial molecules. However, TLRs also recognize some nonmicrobial molecules. In particular, TLR2 and TLR4 recognize hyaluronic acid, a glycosaminoglycan in the extracellular matrix. In neonatal mice endogenous hyaluronic acid binding to TLR4 drives normal intestinal growth. Hyaluronic acid binding to TLR4 in pericryptal macrophages results in cyclooxygenase2- dependent PGE(2) production, which transactivates EGFR in LGR5+ crypt epithelial stem cells leading to increased proliferation. The expanded population of LGR5+ stem cells leads to crypt fission and lengthening of the intestine and colon. Blocking this pathway at any point (TLR4 activation, PGE(2) production, EGFR transactivation) results in diminished intestinal and colonic growth. A similar pathway leads to epithelial proliferation in wound repair. The repair phase of dextran sodium sulfate colitis is marked by increased epithelial proliferation. In this model, TLR2 and TLR4 in pericryptal macrophages are activated by microbial products or by host hyaluronic acid, resulting in production of CXCL12, a chemokine. CXCL12 induces the migration of cyclooxygenase2-expressing mesenchymal stem cells from the lamina propria of the upper colonic crypts to a site adjacent to LGR5+ epithelial stem cells. PGE(2) released by these mesenchymal stem cells transactivates EGFR in LGR5+ epithelial stem cells leading to increased proliferation. Several TLR2 and TLR4 agonists, including hyaluronic acid, are radioprotective in the intestine through the inhibition of radiation-induced apoptosis in LGR5+ epithelial stem cells. Administration of exogenous TLR2 or TLR4 agonists activates TLR2/TLR4 on pericryptal macrophages inducing CXCL12 production with migration of cyclooxygenase2-expressing mesenchymal stem cells from the lamina propria of the villi to a site adjacent to LGR5+ epithelial stem cells. PGE(2) produced by these mesenchymal stem cells, blocks radiation-induced apoptosis in LGR5+ epithelial stem cells by an EGFR mediated pathway. Frontiers Media S.A. 2021-01-21 /pmc/articles/PMC7859088/ /pubmed/33552081 http://dx.doi.org/10.3389/fimmu.2020.617510 Text en Copyright © 2021 Stenson and Ciorba http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Stenson, William F.
Ciorba, Matthew A.
Nonmicrobial Activation of TLRs Controls Intestinal Growth, Wound Repair, and Radioprotection
title Nonmicrobial Activation of TLRs Controls Intestinal Growth, Wound Repair, and Radioprotection
title_full Nonmicrobial Activation of TLRs Controls Intestinal Growth, Wound Repair, and Radioprotection
title_fullStr Nonmicrobial Activation of TLRs Controls Intestinal Growth, Wound Repair, and Radioprotection
title_full_unstemmed Nonmicrobial Activation of TLRs Controls Intestinal Growth, Wound Repair, and Radioprotection
title_short Nonmicrobial Activation of TLRs Controls Intestinal Growth, Wound Repair, and Radioprotection
title_sort nonmicrobial activation of tlrs controls intestinal growth, wound repair, and radioprotection
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7859088/
https://www.ncbi.nlm.nih.gov/pubmed/33552081
http://dx.doi.org/10.3389/fimmu.2020.617510
work_keys_str_mv AT stensonwilliamf nonmicrobialactivationoftlrscontrolsintestinalgrowthwoundrepairandradioprotection
AT ciorbamatthewa nonmicrobialactivationoftlrscontrolsintestinalgrowthwoundrepairandradioprotection