Cargando…

Intestinal-Specific TNFα Overexpression Induces Crohn’s-Like Ileitis in Mice

BACKGROUND AND AIM: Human and animal studies have clearly established tumor necrosis factor (TNF)α as an important mediator of Crohn’s disease pathogenesis. However, whether systemic or only local TNFα overproduction is required for the development of chronic intestinal inflammation and Crohn’s dise...

Descripción completa

Detalles Bibliográficos
Autores principales: Bamias, Giorgos, Dahman, Mohamed I., Arseneau, Kristen O., Guanzon, Mitchell, Gruska, Dennis, Pizarro, Theresa T., Cominelli, Fabio
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3748077/
https://www.ncbi.nlm.nih.gov/pubmed/23977323
http://dx.doi.org/10.1371/journal.pone.0072594
_version_ 1782281030509527040
author Bamias, Giorgos
Dahman, Mohamed I.
Arseneau, Kristen O.
Guanzon, Mitchell
Gruska, Dennis
Pizarro, Theresa T.
Cominelli, Fabio
author_facet Bamias, Giorgos
Dahman, Mohamed I.
Arseneau, Kristen O.
Guanzon, Mitchell
Gruska, Dennis
Pizarro, Theresa T.
Cominelli, Fabio
author_sort Bamias, Giorgos
collection PubMed
description BACKGROUND AND AIM: Human and animal studies have clearly established tumor necrosis factor (TNF)α as an important mediator of Crohn’s disease pathogenesis. However, whether systemic or only local TNFα overproduction is required for the development of chronic intestinal inflammation and Crohn’s disease remains unclear. The aim of this study was to assess the contribution of intestinal epithelial-derived TNFα to the development of murine Crohn’s-like ileitis. METHODS: We adapted the well-established TNF(∆ARE/+) mouse model of Crohn’s disease (which systemically overexpresses TNFα) to generate a homozygous mutant strain that overexpress TNFα only within the intestinal epithelium. Intestinal-specific TNF(i∆ARE/i∆ARE) mice were examined for histopathological signs of gut inflammation and extraintestinal manifestations of Crohn’s disease. The mucosal immune phenotype was characterized, and the contribution of specific lymphocyte populations to the pathogenesis of TNF(i∆ARE/i∆ARE) ileitis was assessed. RESULTS: TNF(i∆ARE/i∆ARE) mice had increased mucosal and systemic TNFα levels compared to wild-type controls (P<0.001), as well as severe chronic ileitis with increased neutrophil infiltration and villous distortion, but no extraintestinal manifestations (P<0.001 vs. wild-type controls). The gut mucosal lymphocytic compartment was also expanded in TNF(i∆ARE/i∆ARE) mice (P<0.05), consisting of activated CD69(+) and CD4(+)CD62L(-) lymphocytes (P<0.05). FasL expression was significantly elevated in the mesenteric lymph nodes of TNF(i∆ARE/i∆ARE) mice (P<0.05). Adoptive transfer of mucosal TNF(i∆ARE/i∆ARE) lymphocytes resulted in ileitis in immunologically naïve severe combined immunodeficiency recipients (P<0.05 vs. wild-type controls), indicating an effector phenotype that was associated with increased production of both Th1 (IFNγ) and Th2 (IL-5, IL-13) cytokines. CONCLUSION: Intestinal epithelial-derived TNFα is sufficient for the induction of Crohn’s-like ileitis, but not for the occurrence of extraintestinal manifestations, in TNF(i∆ARE/i∆ARE) mice. These effects were associated with generation of effector lymphocytes within the intestinal mucosa and dysregulated apoptosis. Thus, targeted intestinal blockade of TNFα may provide an effective means to neutralize gut-derived TNFα with reduced side effects.
format Online
Article
Text
id pubmed-3748077
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-37480772013-08-23 Intestinal-Specific TNFα Overexpression Induces Crohn’s-Like Ileitis in Mice Bamias, Giorgos Dahman, Mohamed I. Arseneau, Kristen O. Guanzon, Mitchell Gruska, Dennis Pizarro, Theresa T. Cominelli, Fabio PLoS One Research Article BACKGROUND AND AIM: Human and animal studies have clearly established tumor necrosis factor (TNF)α as an important mediator of Crohn’s disease pathogenesis. However, whether systemic or only local TNFα overproduction is required for the development of chronic intestinal inflammation and Crohn’s disease remains unclear. The aim of this study was to assess the contribution of intestinal epithelial-derived TNFα to the development of murine Crohn’s-like ileitis. METHODS: We adapted the well-established TNF(∆ARE/+) mouse model of Crohn’s disease (which systemically overexpresses TNFα) to generate a homozygous mutant strain that overexpress TNFα only within the intestinal epithelium. Intestinal-specific TNF(i∆ARE/i∆ARE) mice were examined for histopathological signs of gut inflammation and extraintestinal manifestations of Crohn’s disease. The mucosal immune phenotype was characterized, and the contribution of specific lymphocyte populations to the pathogenesis of TNF(i∆ARE/i∆ARE) ileitis was assessed. RESULTS: TNF(i∆ARE/i∆ARE) mice had increased mucosal and systemic TNFα levels compared to wild-type controls (P<0.001), as well as severe chronic ileitis with increased neutrophil infiltration and villous distortion, but no extraintestinal manifestations (P<0.001 vs. wild-type controls). The gut mucosal lymphocytic compartment was also expanded in TNF(i∆ARE/i∆ARE) mice (P<0.05), consisting of activated CD69(+) and CD4(+)CD62L(-) lymphocytes (P<0.05). FasL expression was significantly elevated in the mesenteric lymph nodes of TNF(i∆ARE/i∆ARE) mice (P<0.05). Adoptive transfer of mucosal TNF(i∆ARE/i∆ARE) lymphocytes resulted in ileitis in immunologically naïve severe combined immunodeficiency recipients (P<0.05 vs. wild-type controls), indicating an effector phenotype that was associated with increased production of both Th1 (IFNγ) and Th2 (IL-5, IL-13) cytokines. CONCLUSION: Intestinal epithelial-derived TNFα is sufficient for the induction of Crohn’s-like ileitis, but not for the occurrence of extraintestinal manifestations, in TNF(i∆ARE/i∆ARE) mice. These effects were associated with generation of effector lymphocytes within the intestinal mucosa and dysregulated apoptosis. Thus, targeted intestinal blockade of TNFα may provide an effective means to neutralize gut-derived TNFα with reduced side effects. Public Library of Science 2013-08-20 /pmc/articles/PMC3748077/ /pubmed/23977323 http://dx.doi.org/10.1371/journal.pone.0072594 Text en © 2013 Bamias et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Bamias, Giorgos
Dahman, Mohamed I.
Arseneau, Kristen O.
Guanzon, Mitchell
Gruska, Dennis
Pizarro, Theresa T.
Cominelli, Fabio
Intestinal-Specific TNFα Overexpression Induces Crohn’s-Like Ileitis in Mice
title Intestinal-Specific TNFα Overexpression Induces Crohn’s-Like Ileitis in Mice
title_full Intestinal-Specific TNFα Overexpression Induces Crohn’s-Like Ileitis in Mice
title_fullStr Intestinal-Specific TNFα Overexpression Induces Crohn’s-Like Ileitis in Mice
title_full_unstemmed Intestinal-Specific TNFα Overexpression Induces Crohn’s-Like Ileitis in Mice
title_short Intestinal-Specific TNFα Overexpression Induces Crohn’s-Like Ileitis in Mice
title_sort intestinal-specific tnfα overexpression induces crohn’s-like ileitis in mice
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3748077/
https://www.ncbi.nlm.nih.gov/pubmed/23977323
http://dx.doi.org/10.1371/journal.pone.0072594
work_keys_str_mv AT bamiasgiorgos intestinalspecifictnfaoverexpressioninducescrohnslikeileitisinmice
AT dahmanmohamedi intestinalspecifictnfaoverexpressioninducescrohnslikeileitisinmice
AT arseneaukristeno intestinalspecifictnfaoverexpressioninducescrohnslikeileitisinmice
AT guanzonmitchell intestinalspecifictnfaoverexpressioninducescrohnslikeileitisinmice
AT gruskadennis intestinalspecifictnfaoverexpressioninducescrohnslikeileitisinmice
AT pizarrotheresat intestinalspecifictnfaoverexpressioninducescrohnslikeileitisinmice
AT cominellifabio intestinalspecifictnfaoverexpressioninducescrohnslikeileitisinmice