Deutsch
 
Hilfe Datenschutzhinweis Impressum
  DetailsucheBrowse

Datensatz

DATENSATZ AKTIONENEXPORT

Freigegeben

Zeitschriftenartikel

Eomesodermin Expression in CD4+

MPG-Autoren

Lupar,  Ekaterina
Max Planck Institute of Immunobiology and Epigenetics, Max Planck Society;
Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg;
Faculty of Biology, University of Freiburg;

Brack,  Maria
Max Planck Institute of Immunobiology and Epigenetics, Max Planck Society;
Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg;

/persons/resource/persons191125

Izcue,  A.
Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Max Planck Society;

Externe Ressourcen
Es sind keine externen Ressourcen hinterlegt
Volltexte (beschränkter Zugriff)
Für Ihren IP-Bereich sind aktuell keine Volltexte freigegeben.
Volltexte (frei zugänglich)
Es sind keine frei zugänglichen Volltexte in PuRe verfügbar
Ergänzendes Material (frei zugänglich)
Es sind keine frei zugänglichen Ergänzenden Materialien verfügbar
Zitation

Lupar, E., Brack, M., Garnier, L., Laffont, S., Rauch, K. S., Schachtrup, K., et al. (2015). Eomesodermin Expression in CD4+. The Journal of Immunology, 195, 4742-4752. doi:doi: 10.4049/jimmunol.1501159.


Zitierlink: https://hdl.handle.net/someHandle/test/escidoc:902521
Zusammenfassung
CD4<sup>+</sup> T cells polarize into effector Th subsets characterized by signature transcription factors and cytokines. Although T-bet drives Th1 responses and represses the alternative Th2, Th17, and Foxp3<sup>+</sup> regulatory T cell fates, the role of the T-bet-related transcription factor eomesodermin (Eomes) in CD4<sup>+</sup> T cells is less well understood. In this study, we analyze the expression and effects of Eomes in mouse CD4<sup>+</sup) T lymphocytes. We find that Eomes is readily expressed in activated CD4<sup>+</sup> Th1 T cells in vivo. Eomes<sup>+</sup> CD4<sup>+</sup> T cells accumulated in old mice, under lymphopenic conditions in a T cell transfer model of colitis, and upon oral Ag administration. However, despite its expression, genetic deletion of Eomes in CD4<sup>+</sup> T cells did not impact on IFN-γ production nor increase Th2 or Th17 responses. In contrast, Eomes deficiency favored the accumulation of Foxp3<sup>+</sup> cells in old mice, after in vivo differentiation of Eomes-deficient naive CD4<sup>+</sup> T cells, and in response to oral Ag in a cell-intrinsic way. Enforced Eomes expression during in vitro regulatory T cell induction also reduced Foxp3 transcription. Likewise, bystander Eomes-deficient CD4<sup>+</sup> T cells were more efficient at protecting from experimental autoimmune encephalitis compared with wild-type CD4<sup>+</sup> T cells. This enhanced capacity of Eomes-deficient CD4<sup>+</sup> T cells to inhibit EAE in trans was associated with an enhanced frequency of Foxp3<sup>+</sup> cells. Our data identify a novel role for Eomes in CD4<sup>+</sup> T cells and indicate that Eomes expression may act by limiting Foxp3 induction, which may contribute to the association of EOMES to susceptibility to multiple sclerosis.