Thymocyte apoptosis drives the intrathymic generation of regulatory T cells

Maintenance of immune tolerance critically depends upon regulatory T cells that express the transcription factor forkhead box P3 (Foxp3). These CD4 ⁺ T cells can be generated in the thymus, termed thymus-derived regulatory T cells (tTregs), but their developmental pathway remains incompletely unders...

Full description

Saved in:
Bibliographic Details
Published inProceedings of the National Academy of Sciences - PNAS Vol. 111; no. 4; pp. E465 - E473
Main Authors Konkel, Joanne E, Jin, Wenwen, Abbatiello, Brittany, Grainger, John R, Chen, WanJun
Format Journal Article
LanguageEnglish
Published United States National Academy of Sciences 28.01.2014
National Acad Sciences
SeriesPNAS Plus
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Maintenance of immune tolerance critically depends upon regulatory T cells that express the transcription factor forkhead box P3 (Foxp3). These CD4 ⁺ T cells can be generated in the thymus, termed thymus-derived regulatory T cells (tTregs), but their developmental pathway remains incompletely understood. tTreg development has been shown to be delayed compared with that of CD4 ⁺ single positive (SP) thymocytes, with tTregs being detected only in neonatal thymi by day 3 after birth. Here, we outline the reasons for this delayed emergence of Foxp3 ⁺ tTregs and demonstrate that thymocyte apoptosis is intrinsically tied to tTreg development. We show that thymic apoptosis leads to the production of TGFβ intrathymically from thymic macrophages, dendritic cells, and epithelial cells. This TGFβ then induces foxp3 expression and drives tTreg generation. Thymocyte apoptosis has previously been shown to accelerate after birth, which drives increases in TGFβ in the neonatal thymus. We highlight a paucity of TGFβ in the neonatal thymus, accounting for the delayed development of tTregs compared with CD4 ⁺ SP thymocytes. Importantly, we show that enhanced levels of apoptosis in the thymus result in an augmented tTreg population and, moreover, that decreasing thymic apoptosis results in reduced tTregs. In addition to this, we also show that T-cell receptor (TCR) signals of different affinity were all capable of driving tTreg development; however, to achieve this TGFβ signals must also be received concomitant with the TCR signal. Collectively, our results indicate that thymic apoptosis is a key event in tTreg generation and reveal a previously unrecognized apoptosis–TGFβ–Foxp3 axis that mediates the development of tTregs.
Bibliography:http://dx.doi.org/10.1073/pnas.1320319111
ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ObjectType-Article-2
ObjectType-Feature-1
Edited by Philippa Marrack, Howard Hughes Medical Institute, National Jewish Health, Denver, CO, and approved December 18, 2013 (received for review October 28, 2013)
Author contributions: J.E.K. and W.C. designed research; J.E.K., W.J., B.A., and J.R.G. performed research; J.E.K. and W.C. analyzed data; and J.E.K. and W.C. wrote the paper.
ISSN:0027-8424
1091-6490
DOI:10.1073/pnas.1320319111