Upon primary exposure to antigen, nave T cells bearing complementary antigen receptors (TCRs) undergo rapid activation and clonal expansion, leading to the generation of effector T cells. The vast majority (90-95%) of antigen-specific effector T cells that participate in the primary immune response undergo apoptosis (cell death) after antigen clearance. A small subset, however, survives and gives rise to long-lived memory T cells. Upon antigen re-encounter, memory T cells respond swiftly and robustly to eliminate the pathogen. Based on their tissue distribution, cell surface markers, and effector functions, memory T cells have been divided into two major subsets. Memory T cells expressing receptors such as CD62L and CCR7, which allow efficient homing to lymph nodes (LN), are termed central memory (TCM) cells;memory T cells lacking LN-homing receptors and preferentially residing in non-lymphoid tissues are termed effector memory (TEM) cells. Notably, both memory subsets display high levels of the marker CD44. Once established CD4 and CD8 memory T cell populations can be maintained for many years in vivo. Although early studies suggested that continued antigenic stimulation is required for maintaining T cell memory, neither cognate antigen nor MHC-encoded molecules appear to be required for the long-term survival of CD4 or CD8 memory T cells. Members of the tumor necrosis factor receptor (TNFR) family are involved in T cell costimulation and play a role in T cell survival. Mice deficient in OX40, CD27, or 4-1BB show greater defects in secondary compared to primary responses when infected with pathogens such as lymphocytic choriomeningitis virus (LCMV), vesicular stomatitis virus, and influenza. Working primarily in mice, we developed a method of separating naive from memory T cells based upon their density. Using this technique, we made the following findings (especially with regard to CD8 T cell memory, which is important in anti-viral and anti-neoplastic responses): 1. Memory T cells exist in the G1 phase (high RNA, haploid DNA) of the cell cycle, whereas naive T cells exist in G0 (low RNA, haploid DNA). Stimulation of the former results in rapid release of high amounts of cytokines such as interferon gamma (IFNgamma). 2. Culture of purified memory T cells in the absence of other cells (resting) allows them to revert to G0. When they do, they respond to stimulation to the same degree and with the same kinetics as naive T cells. 3. Culture of memory T cells with purified dendritic cells (DC) prevents their reversion to G0 and maintains their characteristic potent effector responses. 4. Blockade of two TNFR molecules, CD70 and 4-1BB reverses the """"""""protection"""""""" provided by DC. Furthermore, direct stimulation of the CD70 receptor on T cells, CD27, also maintains T cells in the memory state in the absence of any other cells. 5. All of these results were verified with memory T cells generated against lymphocytic choriomeningitis virus (LCMV) and identified by MHC-peptide """"""""tetramers"""""""", which can identify individual antigen-specific cells. 6. CD27-deficient mice, memory T cells responsive to vesicular stomatitis virus (VSV) have difficulty maintaining their G1 state. 7. Ligation of CD27 activated phosphatidylinositol-3 kinase (PI3K) and Akt. This in turn resulted in phosphorylation of the quiescence transcription factor FOXO1 and its export from the nucleus (deactivation). Notably, whereas freshly isolated naive T cells expressed a substantial amount of FOXO1, levels were markedly reduced in memory T cells. These results account for the requirement for CD27 signaling in memory T cell maintenance. PI3K/Akt activate mammalian target of rapamycin (mTOR), which plays a vital role in protein translation and cell cycle progression. FOXO1 is important for maintaining cells in a quiescent state (i.e. in G0). Based upon this work, we have generated CD70-deficient mice. We have found that when normal memory T cells are adoptively transferred to CD70-deficient mice, they lose the ability to respond to antigenic rechallenge. The mechanistic basis for this phenomenon is under investigation.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Investigator-Initiated Intramural Research Projects (ZIA)
Project #
1ZIABC010932-04
Application #
8349235
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
4
Fiscal Year
2011
Total Cost
$422,756
Indirect Cost
Name
National Cancer Institute Division of Basic Sciences
Department
Type
DUNS #
City
State
Country
Zip Code
Allam, Atef; Swiecki, Melissa; Vermi, William et al. (2014) Dual function of CD70 in viral infection: modulator of early cytokine responses and activator of adaptive responses. J Immunol 193:871-8
Kuka, Mirela; Munitic, Ivana; Giardino Torchia, Maria Letizia et al. (2013) CD70 is downregulated by interaction with CD27. J Immunol 191:2282-9
Munitic, Ivana; Kuka, Mirela; Allam, Atef et al. (2013) CD70 deficiency impairs effector CD8 T cell generation and viral clearance but is dispensable for the recall response to lymphocytic choriomeningitis virus. J Immunol 190:1169-79
Kuka, Mirela; Ashwell, Jonathan D (2013) A method for high purity sorting of rare cell subsets applied to TDC. J Immunol Methods 400-401:111-6
Kuka, Mirela; Munitic, Ivana; Ashwell, Jonathan D (2012) Identification and characterization of polyclonal ??-T cells with dendritic cell properties. Nat Commun 3:1223
Allam, Atef; Conze, Dietrich B; Giardino Torchia, Maria Letizia et al. (2009) The CD8+ memory T-cell state of readiness is actively maintained and reversible. Blood 114:2121-30