TNF receptor type 2 (p75) functions as a costimulator for antigen-driven T cell responses in vivo

EY Kim, JJ Priatel, SJ Teh, HS Teh - The Journal of Immunology, 2006 - journals.aai.org
EY Kim, JJ Priatel, SJ Teh, HS Teh
The Journal of Immunology, 2006journals.aai.org
Naive T cells require costimulation for robust Ag-driven differentiation and survival. Members
of the TNFR family have been shown to provide costimulatory signals conferring survival at
distinct phases of the T cell response. In this study, we show that CD4 and CD8 T cells
depend on TNFR type 2 (p75) for survival during clonal expansion, allowing larger
accumulation of effector cells and conferring protection from apoptosis for a robust memory
pool in vivo. We demonstrate using the MHC class I-restricted 2C TCR and MHC class II …
Abstract
Naive T cells require costimulation for robust Ag-driven differentiation and survival. Members of the TNFR family have been shown to provide costimulatory signals conferring survival at distinct phases of the T cell response. In this study, we show that CD4 and CD8 T cells depend on TNFR type 2 (p75) for survival during clonal expansion, allowing larger accumulation of effector cells and conferring protection from apoptosis for a robust memory pool in vivo. We demonstrate using the MHC class I-restricted 2C TCR and MHC class II-restricted AND TCR transgenic systems that TNFR2 regulates the threshold for clonal expansion of CD4 and CD8 T cell subsets in response to cognate Ag. Using a novel recombinant Listeria monocytogenes (rLM) expressing a secreted form of the 2C agonist peptide (SIY) to investigate the role of TNFR2 for T cell immunity in vivo, we found that TNFR2 controls the survival and accumulation of effector cells during the primary response. TNFR2−/− CD8 T cells exhibit loss of protection from apoptosis that is correlated with diminished survivin and Bcl-2 expression. Null mutant mice were more susceptible to rLM-SIY challenge at high doses of primary infection, correlating with impaired LM-specific T cell response in the absence of TNFR2-mediated costimulation. Moreover, the resulting memory pools specific for SIY and listeriolysin O epitopes derived from rLM-SIY were diminished in TNFR2−/− mice. Thus, examination of Ag-driven T cell responses revealed a hitherto unknown costimulatory function for TNFR2 in regulating T cell survival during the differentiation program elicited by intracellular pathogen in vivo.
journals.aai.org