A convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector T cells and without perturbation of regulatory T cells.

Autoimmune gastritis results from the breakdown of T cell tolerance to the gastric H(+)/K(+) ATPase. The gastric H(+)/K(+) ATPase is responsible for the acidification of gastric juice and consists of an α subunit (H/Kα) and a β subunit (H/Kβ). Here we show that CD4(+) T cells from H/Kα-deficient mic...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Eric Tu, Desmond K Y Ang, Thea V Hogan, Simon Read, Cheryl P Z Chia, Paul A Gleeson, Ian R van Driel
Formato: article
Lenguaje:EN
Publicado: Public Library of Science (PLoS) 2011
Materias:
R
Q
Acceso en línea:https://doaj.org/article/0b44e4837783435d8d40abcf6bc5c378
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:0b44e4837783435d8d40abcf6bc5c378
record_format dspace
spelling oai:doaj.org-article:0b44e4837783435d8d40abcf6bc5c3782021-11-18T07:34:39ZA convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector T cells and without perturbation of regulatory T cells.1932-620310.1371/journal.pone.0027153https://doaj.org/article/0b44e4837783435d8d40abcf6bc5c3782011-01-01T00:00:00Zhttps://www.ncbi.nlm.nih.gov/pmc/articles/pmid/22096532/?tool=EBIhttps://doaj.org/toc/1932-6203Autoimmune gastritis results from the breakdown of T cell tolerance to the gastric H(+)/K(+) ATPase. The gastric H(+)/K(+) ATPase is responsible for the acidification of gastric juice and consists of an α subunit (H/Kα) and a β subunit (H/Kβ). Here we show that CD4(+) T cells from H/Kα-deficient mice (H/Kα(-/-)) are highly pathogenic and autoimmune gastritis can be induced in sublethally irradiated wildtype mice by adoptive transfer of unfractionated CD4(+) T cells from H/Kα(-/-) mice. All recipient mice consistently developed the most severe form of autoimmune gastritis 8 weeks after the transfer, featuring hypertrophy of the gastric mucosa, complete depletion of the parietal and zymogenic cells, and presence of autoantibodies to H(+)/K(+) ATPase in the serum. Furthermore, we demonstrated that the disease significantly affected stomach weight and stomach pH of recipient mice. Depletion of parietal cells in this disease model required the presence of both H/Kα and H/Kβ since transfer of H/Kα(-/-) CD4(+) T cells did not result in depletion of parietal cells in H/Kα(-/-) or H/Kβ(-/-) recipient mice. The consistency of disease severity, the use of polyclonal T cells and a specific T cell response to the gastric autoantigen make this an ideal disease model for the study of many aspects of organ-specific autoimmunity including prevention and treatment of the disease.Eric TuDesmond K Y AngThea V HoganSimon ReadCheryl P Z ChiaPaul A GleesonIan R van DrielPublic Library of Science (PLoS)articleMedicineRScienceQENPLoS ONE, Vol 6, Iss 11, p e27153 (2011)
institution DOAJ
collection DOAJ
language EN
topic Medicine
R
Science
Q
spellingShingle Medicine
R
Science
Q
Eric Tu
Desmond K Y Ang
Thea V Hogan
Simon Read
Cheryl P Z Chia
Paul A Gleeson
Ian R van Driel
A convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector T cells and without perturbation of regulatory T cells.
description Autoimmune gastritis results from the breakdown of T cell tolerance to the gastric H(+)/K(+) ATPase. The gastric H(+)/K(+) ATPase is responsible for the acidification of gastric juice and consists of an α subunit (H/Kα) and a β subunit (H/Kβ). Here we show that CD4(+) T cells from H/Kα-deficient mice (H/Kα(-/-)) are highly pathogenic and autoimmune gastritis can be induced in sublethally irradiated wildtype mice by adoptive transfer of unfractionated CD4(+) T cells from H/Kα(-/-) mice. All recipient mice consistently developed the most severe form of autoimmune gastritis 8 weeks after the transfer, featuring hypertrophy of the gastric mucosa, complete depletion of the parietal and zymogenic cells, and presence of autoantibodies to H(+)/K(+) ATPase in the serum. Furthermore, we demonstrated that the disease significantly affected stomach weight and stomach pH of recipient mice. Depletion of parietal cells in this disease model required the presence of both H/Kα and H/Kβ since transfer of H/Kα(-/-) CD4(+) T cells did not result in depletion of parietal cells in H/Kα(-/-) or H/Kβ(-/-) recipient mice. The consistency of disease severity, the use of polyclonal T cells and a specific T cell response to the gastric autoantigen make this an ideal disease model for the study of many aspects of organ-specific autoimmunity including prevention and treatment of the disease.
format article
author Eric Tu
Desmond K Y Ang
Thea V Hogan
Simon Read
Cheryl P Z Chia
Paul A Gleeson
Ian R van Driel
author_facet Eric Tu
Desmond K Y Ang
Thea V Hogan
Simon Read
Cheryl P Z Chia
Paul A Gleeson
Ian R van Driel
author_sort Eric Tu
title A convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector T cells and without perturbation of regulatory T cells.
title_short A convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector T cells and without perturbation of regulatory T cells.
title_full A convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector T cells and without perturbation of regulatory T cells.
title_fullStr A convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector T cells and without perturbation of regulatory T cells.
title_full_unstemmed A convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector T cells and without perturbation of regulatory T cells.
title_sort convenient model of severe, high incidence autoimmune gastritis caused by polyclonal effector t cells and without perturbation of regulatory t cells.
publisher Public Library of Science (PLoS)
publishDate 2011
url https://doaj.org/article/0b44e4837783435d8d40abcf6bc5c378
work_keys_str_mv AT erictu aconvenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT desmondkyang aconvenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT theavhogan aconvenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT simonread aconvenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT cherylpzchia aconvenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT paulagleeson aconvenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT ianrvandriel aconvenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT erictu convenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT desmondkyang convenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT theavhogan convenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT simonread convenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT cherylpzchia convenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT paulagleeson convenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
AT ianrvandriel convenientmodelofseverehighincidenceautoimmunegastritiscausedbypolyclonaleffectortcellsandwithoutperturbationofregulatorytcells
_version_ 1718423203680878592