NK cells delay allograft rejection in lymphopenic hosts by downregulating the homeostatic proliferation of CD8+ T cells

D Zecher, Q Li, MH Oberbarnscheidt… - The Journal of …, 2010 - journals.aai.org
D Zecher, Q Li, MH Oberbarnscheidt, AJ Demetris, WD Shlomchik, DM Rothstein, FG Lakkis
The Journal of Immunology, 2010journals.aai.org
T cells present in lymphopenic environments undergo spontaneous (homeostatic)
proliferation resulting in expansion of the memory T cell pool. Homeostatically generated
memory T cells protect the host against infection but can cause autoimmunity and allograft
rejection. Therefore, understanding the mechanisms that regulate homeostatic T cell
proliferation is germane to clinical settings in which lymphodepletion is used. In this study,
we asked whether NK cells, which regulate immune responses in lymphocyte-replete hosts …
Abstract
T cells present in lymphopenic environments undergo spontaneous (homeostatic) proliferation resulting in expansion of the memory T cell pool. Homeostatically generated memory T cells protect the host against infection but can cause autoimmunity and allograft rejection. Therefore, understanding the mechanisms that regulate homeostatic T cell proliferation is germane to clinical settings in which lymphodepletion is used. In this study, we asked whether NK cells, which regulate immune responses in lymphocyte-replete hosts, also regulate homeostatic T cell proliferation under lymphopenic conditions. We found that T cells transferred into genetically lymphocyte-deficient RAG−/− mice proliferate faster and generate more CD8+ memory T cells if NK cells were absent. CD8+ T cells that underwent homeostatic proliferation in the presence of NK cells generated mostly effector memory (CD44 high CD62L low) lymphocytes, whereas those that divided in the absence of NK cells were skewed toward central memory (CD44 high CD62L high). The latter originated predominantly from proliferation of the “natural” central memory CD8+ T cell pool. Regulation of homeostatic proliferation by NK cells occurred independent of perforin but was reversed by excess IL-15. Importantly, NK depletion enhanced CD8+ T cell recovery in T cell-depleted wild-type mice and accelerated rejection of skin allografts, indicating that regulation of homeostatic proliferation by NK cells is not restricted to genetically lymphocyte-deficient animals. These results demonstrate that NK cells downregulate homeostatic CD8+ T cell proliferation in lymphopenic environments by competing for IL-15. Concomitant NK and T cell depletion may be undesirable in transplant recipients because of enhanced expansion of memory CD8+ T cells that increase the risk of rejection.
journals.aai.org