Small intestine inflammation in Roquin-mutant and Roquin-deficient mice.

Roquin, an E3 ubiquitin ligase that localizes to cytosolic RNA granules, is involved in regulating mRNA stability and translation. Mice that have a M199R mutation in the Roquin protein (referred to as sanroque or Roquin(san/san) mice) develop autoimmune pathologies, although the extent to which thes...

Full description

Bibliographic Details
Main Authors: Jeremy S Schaefer, Dina Montufar-Solis, Niyati Nakra, Nadarajah Vigneswaran, John R Klein
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2013-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3581552?pdf=render
id doaj-accfff4cd7704b65bc4cee5a443a83c1
record_format Article
spelling doaj-accfff4cd7704b65bc4cee5a443a83c12020-11-25T01:45:20ZengPublic Library of Science (PLoS)PLoS ONE1932-62032013-01-0182e5643610.1371/journal.pone.0056436Small intestine inflammation in Roquin-mutant and Roquin-deficient mice.Jeremy S SchaeferDina Montufar-SolisNiyati NakraNadarajah VigneswaranJohn R KleinRoquin, an E3 ubiquitin ligase that localizes to cytosolic RNA granules, is involved in regulating mRNA stability and translation. Mice that have a M199R mutation in the Roquin protein (referred to as sanroque or Roquin(san/san) mice) develop autoimmune pathologies, although the extent to which these occur in the intestinal mucosa has not been determined. Here, we demonstrate that Roquin(san/san) mice reproducibly develop intestinal inflammation in the small intestine but not the colon. Similarly, mice generated in our laboratory in which the Roquin gene was disrupted by insertion of a gene trap cassette (Roquin(gt/gt) mice) had small intestinal inflammation that mimicked that of Roquin(san/san) mice. MLN cells in Roquin(san/san) mice consisted of activated proliferating T cells, and had increased numbers of CD44(hi) CD62L(lo) KLRG1(+) short-lived effector cells. Proportionally more small intestinal intraepithelial lymphocytes in Roquin(san/san) mice expressed the ICOS T cell activation marker. Of particular interest, small intestinal lamina propria lymphocytes in Roquin(san/san) mice consisted of a high proportion of Gr-1(+) T cells that included IL-17A(+) cells and CD8(+) IFN-γ(+) cells. Extensive cytokine dysregulation resulting in both over-expression and under-expression of chemotactic cytokines occurred in the ileum of Roquin(san/san) mice, the region most prone to the development of inflammation. These findings demonstrate that chronic inflammation ensues in the intestine following Roquin alteration either as a consequence of protein mutation or gene disruption, and they have implications for understanding how small intestinal inflammation is perpetuated in Crohn's disease (CD). Due to the paucity of animal models of CD-like pathophysiology in the small intestine, and because the primary gene/protein defects of the Roquin animal systems used here are well-defined, it will be possible to further elucidate the underlying genetic and molecular mechanisms that drive the disease process.http://europepmc.org/articles/PMC3581552?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Jeremy S Schaefer
Dina Montufar-Solis
Niyati Nakra
Nadarajah Vigneswaran
John R Klein
spellingShingle Jeremy S Schaefer
Dina Montufar-Solis
Niyati Nakra
Nadarajah Vigneswaran
John R Klein
Small intestine inflammation in Roquin-mutant and Roquin-deficient mice.
PLoS ONE
author_facet Jeremy S Schaefer
Dina Montufar-Solis
Niyati Nakra
Nadarajah Vigneswaran
John R Klein
author_sort Jeremy S Schaefer
title Small intestine inflammation in Roquin-mutant and Roquin-deficient mice.
title_short Small intestine inflammation in Roquin-mutant and Roquin-deficient mice.
title_full Small intestine inflammation in Roquin-mutant and Roquin-deficient mice.
title_fullStr Small intestine inflammation in Roquin-mutant and Roquin-deficient mice.
title_full_unstemmed Small intestine inflammation in Roquin-mutant and Roquin-deficient mice.
title_sort small intestine inflammation in roquin-mutant and roquin-deficient mice.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2013-01-01
description Roquin, an E3 ubiquitin ligase that localizes to cytosolic RNA granules, is involved in regulating mRNA stability and translation. Mice that have a M199R mutation in the Roquin protein (referred to as sanroque or Roquin(san/san) mice) develop autoimmune pathologies, although the extent to which these occur in the intestinal mucosa has not been determined. Here, we demonstrate that Roquin(san/san) mice reproducibly develop intestinal inflammation in the small intestine but not the colon. Similarly, mice generated in our laboratory in which the Roquin gene was disrupted by insertion of a gene trap cassette (Roquin(gt/gt) mice) had small intestinal inflammation that mimicked that of Roquin(san/san) mice. MLN cells in Roquin(san/san) mice consisted of activated proliferating T cells, and had increased numbers of CD44(hi) CD62L(lo) KLRG1(+) short-lived effector cells. Proportionally more small intestinal intraepithelial lymphocytes in Roquin(san/san) mice expressed the ICOS T cell activation marker. Of particular interest, small intestinal lamina propria lymphocytes in Roquin(san/san) mice consisted of a high proportion of Gr-1(+) T cells that included IL-17A(+) cells and CD8(+) IFN-γ(+) cells. Extensive cytokine dysregulation resulting in both over-expression and under-expression of chemotactic cytokines occurred in the ileum of Roquin(san/san) mice, the region most prone to the development of inflammation. These findings demonstrate that chronic inflammation ensues in the intestine following Roquin alteration either as a consequence of protein mutation or gene disruption, and they have implications for understanding how small intestinal inflammation is perpetuated in Crohn's disease (CD). Due to the paucity of animal models of CD-like pathophysiology in the small intestine, and because the primary gene/protein defects of the Roquin animal systems used here are well-defined, it will be possible to further elucidate the underlying genetic and molecular mechanisms that drive the disease process.
url http://europepmc.org/articles/PMC3581552?pdf=render
work_keys_str_mv AT jeremysschaefer smallintestineinflammationinroquinmutantandroquindeficientmice
AT dinamontufarsolis smallintestineinflammationinroquinmutantandroquindeficientmice
AT niyatinakra smallintestineinflammationinroquinmutantandroquindeficientmice
AT nadarajahvigneswaran smallintestineinflammationinroquinmutantandroquindeficientmice
AT johnrklein smallintestineinflammationinroquinmutantandroquindeficientmice
_version_ 1725023528030306304