scholarly journals CD8 T Cell Memory Recall Is Enhanced by Novel Direct Interactions with CD4 T Cells Enabled by MHC Class II Transferred from APCs

PLoS ONE ◽  
2013 ◽  
Vol 8 (2) ◽  
pp. e56999 ◽  
Author(s):  
Pablo A. Romagnoli ◽  
Mary F. Premenko-Lanier ◽  
Gilbert D. Loria ◽  
John D. Altman
2006 ◽  
Vol 176 (4) ◽  
pp. 2486-2495 ◽  
Author(s):  
Qigui Yu ◽  
Feng Yun Yue ◽  
Xiao X. Gu ◽  
Herbert Schwartz ◽  
Colin M. Kovacs ◽  
...  

2003 ◽  
Vol 33 (12) ◽  
pp. 3225-3231 ◽  
Author(s):  
Christine Bourgeois ◽  
Corinne Tanchot

2012 ◽  
Vol 18 (3) ◽  
pp. 422-428 ◽  
Author(s):  
Andrew Zloza ◽  
Frederick J Kohlhapp ◽  
Gretchen E Lyons ◽  
Jason M Schenkel ◽  
Tamson V Moore ◽  
...  

2016 ◽  
Vol 16 (2) ◽  
pp. 102-111 ◽  
Author(s):  
Brian J. Laidlaw ◽  
Joseph E. Craft ◽  
Susan M. Kaech

2013 ◽  
Vol 210 (7) ◽  
pp. 1463-1479 ◽  
Author(s):  
Salvador Iborra ◽  
Manuel Ramos ◽  
David M. Arana ◽  
Silvia Lázaro ◽  
Francisco Aguilar ◽  
...  

Signals from the TCR that specifically contribute to effector versus memory CD8+ T cell differentiation are poorly understood. Using mice and adoptively transferred T lymphocytes lacking the small GTPase N-ras, we found that N-ras–deficient CD8+ T cells differentiate efficiently into antiviral primary effectors but have a severe defect in generating protective memory cells. This defect was rescued, although only partly, by rapamycin-mediated inhibition of mammalian target of rapamycin (mTOR) in vivo. The memory defect correlated with a marked impairment in vitro and in vivo of the antigen-mediated early induction of T-box transcription factor Eomesodermin (Eomes), whereas T-bet was unaffected. Besides N-ras, early Eomes induction in vitro required phosphoinositide 3-kinase (PI3K)–AKT but not extracellular signal-regulated kinase (ERK) activation, and it was largely insensitive to rapamycin. Consistent with N-ras coupling Eomes to T cell memory, retrovirally enforced expression of Eomes in N-ras–deficient CD8+ T cells effectively rescued their memory differentiation. Thus, our study identifies a critical role for N-ras as a TCR-proximal regulator of Eomes for early determination of the CD8+ T cell memory fate.


2013 ◽  
Vol 74 (7) ◽  
pp. 809-817 ◽  
Author(s):  
Vivian Zhou ◽  
Maryam B. Yassai ◽  
Jeyarani Regunathan ◽  
Jodie Box ◽  
Dmitry Bosenko ◽  
...  

2017 ◽  
Vol 114 (9) ◽  
pp. E1659-E1667 ◽  
Author(s):  
Karin M. Knudson ◽  
Curtis J. Pritzl ◽  
Vikas Saxena ◽  
Amnon Altman ◽  
Mark A. Daniels ◽  
...  

T-cell memory is critical for long-term immunity. However, the factors involved in maintaining the persistence, function, and phenotype of the memory pool are undefined. Eomesodermin (Eomes) is required for the establishment of the memory pool. Here, we show that in T cells transitioning to memory, the expression of high levels of Eomes is not constitutive but rather requires a continuum of cell-intrinsic NFκB signaling. Failure to maintain NFκB signals after the peak of the response led to impaired Eomes expression and a defect in the maintenance of CD8 T-cell memory. Strikingly, we found that antigen receptor [T-cell receptor (TCR)] signaling regulates this process through expression of the NFκB-dependent kinase proviral integration site for Moloney murine leukemia virus-1 (PIM-1), which in turn regulates NFκB and Eomes. T cells defective in TCR-dependent NFκB signaling were impaired in late expression of Pim-1, Eomes, and CD8 memory. These defects were rescued when TCR-dependent NFκB signaling was restored. We also found that NFκB–Pim-1 signals were required at memory to maintain memory CD8 T-cell longevity, effector function, and Eomes expression. Hence, an NFκB–Pim-1–Eomes axis regulates Eomes levels to maintain memory fitness.


2015 ◽  
Vol 75 (18) ◽  
pp. 3747-3759 ◽  
Author(s):  
Marco Donia ◽  
Rikke Andersen ◽  
Julie W. Kjeldsen ◽  
Paolo Fagone ◽  
Shamaila Munir ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document