scholarly journals Tumor Progression Locus 2 (Tpl2) Activates the Mammalian Target of Rapamycin (mTOR) Pathway, Inhibits Forkhead Box P3 (FoxP3) Expression, and Limits Regulatory T Cell (Treg) Immunosuppressive Functions

2016 ◽  
Vol 291 (32) ◽  
pp. 16802-16815 ◽  
Author(s):  
Xin Li ◽  
Nicole V. Acuff ◽  
Angela R. Peeks ◽  
Rebecca Kirkland ◽  
Kara D. Wyatt ◽  
...  
Hepatology ◽  
2010 ◽  
Vol 51 (5) ◽  
pp. 1789-1798 ◽  
Author(s):  
Pascal Lapierre ◽  
Kathie Béland ◽  
Caroline Martin ◽  
Fernando Alvarez ◽  
Fernando Alvarez

Blood ◽  
2010 ◽  
Vol 115 (23) ◽  
pp. 4758-4769 ◽  
Author(s):  
Hēth R. Turnquist ◽  
Jon Cardinal ◽  
Camila Macedo ◽  
Brian R. Rosborough ◽  
Tina L. Sumpter ◽  
...  

Abstract Prolonged inhibition of the kinase, mammalian target of rapamycin (mTOR), during myeloid dendritic cell (DC) generation confers resistance to maturation. Recently, however, mTOR inhibition immediately before Toll-like receptor ligation has been found to exert proinflammatory effects on myeloid cells, notably enhanced IL-12p40/p70 production. We show, for the first time, that mouse or human DCs generated under mTOR inhibition exhibit markedly enhanced IL-12p70 production after lipopolysaccharide (LPS) stimulation, despite impaired costimulatory molecule expression and poor T-cell stimulatory ability. Consistent with this finding, we reveal that increased IL-12p40 production occurs predominantly in CD86lo immature DCs. High IL-12p40/p70 production by CD86lo DC resulted from failed down-regulation of glycogen synthase kinase-3 (GSK-3) activity and could not be ascribed to enhanced Akt function. Despite high IL-12p70 secretion, rapamycin-conditioned, LPS-stimulated DCs remained poor T-cell stimulators, failing to enhance allogeneic Th1 cell responses. We also report that inhibition of GSK-3 impedes the ability of LPS-stimulated DCs to induce forkhead box p3 in CD4+CD25− T cells, as does the absence of IL-12p40/p70. Thus, GSK-3 activity in DC is regulated via signaling linked to mTOR and modulates their capacity both to produce IL-12p40/p70 and induce forkhead box p3 in CD4+ T cells under inflammatory conditions.


2008 ◽  
Vol 152 (1) ◽  
pp. 127-137 ◽  
Author(s):  
F. Idali ◽  
J. Wahlström ◽  
C. Müller-Suur ◽  
A. Eklund ◽  
J. Grunewald

Blood ◽  
2008 ◽  
Vol 111 (1) ◽  
pp. 453-462 ◽  
Author(s):  
Robert Zeiser ◽  
Dennis B. Leveson-Gower ◽  
Elizabeth A. Zambricki ◽  
Neeraja Kambham ◽  
Andreas Beilhack ◽  
...  

Based on their ability to control T-cell homeostasis, Foxp3+CD4+CD25+ regulatory T cells (Tregs) are being considered for treatment of autoimmune disorders and acute graft-versus-host disease (aGVHD). When combining Tregs with the immunosuppressant rapamycin (RAPA), we observed reduced alloreactive conventional T-cell (Tconv) expansion and aGVHD lethality compared with each treatment alone. This synergistic in vivo protection was paralleled by intact expansion of polyclonal Tregs with conserved high FoxP3 expression. In contrast to Tconv, activation of Tregs with alloantigen and interleukin-2 preferentially led to signal transducer and activator of transcription 5 (STAT5) phosphorylation and not phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway activity. Expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a negative regulator of the PI3K/Akt/mTOR pathway, remained high in Tregs but not Tconv during stimulation. Conversely, targeted deletion of PTEN increased susceptibility of Tregs to mTOR inhibition by RAPA. Differential impact of RAPA as a result of reduced usage of the mTOR pathway in Tregs compared with conventional T cells explains the synergistic effect of RAPA and Tregs in aGVHD protection, which has important implications for clinical trials using Tregs.


Sign in / Sign up

Export Citation Format

Share Document