Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2008 Aug;8(8):1652-61.
doi: 10.1111/j.1600-6143.2008.02302.x. Epub 2008 Jun 18.

Donor-reactive CD8 memory T cells infiltrate cardiac allografts within 24-h posttransplant in naive recipients

Affiliations

Donor-reactive CD8 memory T cells infiltrate cardiac allografts within 24-h posttransplant in naive recipients

A D Schenk et al. Am J Transplant. 2008 Aug.

Abstract

Normal immune responses stimulated by pathogenic and environmental antigens generate memory T cells that react with donor antigens and no currently used immunosuppressive drug completely inhibits memory T-cell function. While donor-reactive memory T cells clearly compromise graft outcomes, mechanisms utilized by memory T cells to promote rejection are largely unknown. In this study, we investigated how early endogenous memory cells infiltrate and express effector function in cardiac allografts. Endogenous CD8 memory T cells in nonsensitized recipients distinguish syngeneic versus allogeneic cardiac allografts within 24 h of reperfusion. CD8-dependent production of IFN-gamma and CXCL9/Mig was observed 24 to 72 h posttransplant in allografts but not isografts. CXCL9 was produced by donor cells in response to IFN-gamma made by recipient CD8 T cells reactive to donor class I major histocompatibility complex (MHC) molecules. Activated CD8 T cells were detected in allografts at least 3 days before donor-specific effector T cells producing IFN-gamma were detected in the recipient spleen. Early inflammation mediated by donor-reactive CD8 memory T cells greatly enhanced primed effector T-cell infiltration into allografts. These results suggest that strategies for optimal inhibition of alloimmunity should include neutralization of infiltrating CD8 memory T cells within a very narrow window after transplantation.

PubMed Disclaimer

Figures

Figure 1
Figure 1
Early induction of IFN-γ and CXCL9 in allografts is CD8 T cell dependent. A. C57BL/6 mice received syngeneic or complete MHC mismatched A/J cardiac allografts. Groups of 4 iso- (—) and allografts ( - - ) were harvested 24, 48, and 72 hours post-transplant, RNA was isolated from total graft homogenates, and relative real-time PCR was used to measure cytokine and chemokine expression. Data are normalized relative to isograft expression levels at 24 hours post-transplant. ( formula image = p ≤ 0.05) B. Wild-type C57BL/6, B6.CD4−/−, B6.CD8−/− or B6.Rag1−/− mice received syngeneic or A/J cardiac allografts. Groups of 5 grafts were harvested 72 hours post-transplant and intragraft levels of CXCL9 were measured by ELISA. ( formula image = p ≤ 0.0001, formula image = p ≤ 0.002)
Figure 2
Figure 2
Early CXCL9 production requires IFN-γ produced by class I MHC alloreactive CD8 T cells. A. Ld-reactive 2C TCR-transgenic CD8 cells or IFN-γ−/− 2C cells were adoptively transferred into Rag1−/− mice. After 10 weeks BALB/C (Ld-expressing) or DBA/1 (Ld null) hearts were transplanted into groups (n = 4) of reconstituted Rag1−/− recipients and graft CXCL9 proteins levels were measured by ELISA 72 hours post-transplant. ( formula image = p ≤ 0.05 vs. BALB/C ≤ Rag1−/− + 2C) B. (C57BL/10xDBA/1)F1 mice received syngeneic, class II MHC-disparate bq1, or complete MHC-disparate C3H cardiac allografts. Protein levels of CXCL9 were measured in groups of 5 grafts harvested 72 hours after transplantation. ( formula image = p ≤ 0.005 vs. complete MHC disparity)
Figure 3
Figure 3
Donor cells are the major source of CXCL9 produced in the graft early post-transplant. Wild-type, B6.CXCL9−/−, or B6.IFN-γR−/− mice received wild-type syngeneic, wild-type A/J, or A/J.CXCL9−/− cardiac allografts. Protein levels of CXCL9 were measured in groups of 5 grafts harvested 72 hours after transplantation. ( formula image = p ≤ 0.01)
Figure 4
Figure 4
Activated CD8 T cells infiltrate allografts before donor-specific T cell priming is detected in the spleen. A. Flow cytometry was used to detect C57BL/6 CD8 T cells infiltrating A/J cardiac allografts 24, 48, and 72 hours post-transplant. Representative data from 3 independent experiments are shown. B. Cardiac grafts and spleens were removed from C57BL/6 recipients of A/J allografts 48 hours after transplantation. Equal numbers of splenic and graft-infiltrating CD8 cells were isolated by flow sorting and stained for further analysis. Representative data are shown. C. Spleens were harvested from non-transplanted mice or C57BL/6 recipients of A/J cardiac allografts on days 1–5 post-transplant. Splenocytes were mixed with A/J (■) or third party DBA/1 (□) stimulator cells in 24-hour IFN-γ ELISPOT assays. (n = 3/group, formula image = p ≤ 0.05, formula image = p ≤ 0.005)
Figure 5
Figure 5
Adoptive transfer of CD8+CD44hi memory cells restores early CXCL9 production in CD8−/− allograft recipients. A/J skin grafts were placed on C57BL/6 mice and 8–10 weeks later naïve (CD8+CD44lo) and memory (CD8+CD44hi) cell populations were flow-sort purified from spleens and 5 × 106 cells were adoptively transferred to B6.CD8−/− mice which then received either A/J or DBA/1 cardiac allografts. For adoptive transfer of primed T cells, CD8 T cells were isolated from spleens of C57BL/6 mice actively rejecting A/J cardiac allografts at day 7 post-transplant. A. Flow cytometric detection of adoptively transferred CD8 T cells infiltrating allografts in CD8−/− recipients 72 hours post-transplant. Representative data are shown. B. Quantification of the adoptively transferred CD8 T cell populations within spleens and allografts 72 hours post-transplant (n = 4/group, formula image = p ≤ 0.05). C. Quantification of IFN-γ and CXCL9 mRNA within allografts 72 hours post-transplant and normalized to expression levels in the no-transfer control group (n = 4/group, formula image = p ≤ 0.05). D. Using the experimental design described above, 2 × 105 central memory (CD8+CD44hiCD62Lhi) or effector memory (CD8+CD44hiCD62Llo) cells were flow-sort purified and adoptively transferred into CD8−/− recipients of A/J cardiac allografts. Quantification of graft-infiltration by the adoptively transferred CD8 T cells 72 hours after transplantation is shown (n = 4/group).
Figure 6
Figure 6
Early CD8 T cell alloreactivity is not inhibited by α-CD40L mAb or CTLA4-Ig. C57BL/6 recipients of A/J cardiac allografts were treated with control Ig or α-CD40L mAb (0.4 mg on day 0, +1), CTLA4-Ig (0.35 mg on day 0, +1, +2), or α-IFN-γ (0.2 mg 8 hours before and after transplantation). Protein levels of CXCL9 were measured in grafts harvested 72 hours after transplantation (n ≥ 3 group and the experiment was performed twice with identical results, formula image = p ≤ 0.05 vs. IgG).
Figure 7
Figure 7
Early CD8 memory T cell alloreactivity enhances recruitment of primed effector T cells to the allograft. Wild-type C57BL/6 mice were treated with control Ig or CD8 depleting antibodies (0.2 mg on days −3, −2, −1, +1). On day 0 mice received A/J cardiac allografts. On day +2, 8 × 106 purified CD90.1 CD4+ cells from spleens of wild-type CD90.1 C57BL/6 mice rejecting A/J cardiac allografts on day 7 post-transplant were adoptively transferred and 48 hours later allografts were harvested and graft-infiltrating CD90.1 CD4+ T cells were detected using flow cytometry. A. Representative data illustrating the gating strategy used to identify the graft-infiltrating CD4+ CD90.1 test population. B. Quantification of the total number of graft-infiltrating CD4+ CD90.1 cells (n = 6/group, formula image = p ≤ 0.05). C–D. Relative mRNA quantification of IFN-γ and CXCL9 in grafts harvested from IgG and γ-CD8 treated recipients 4 days after transplantation and 48 hours after adoptive transfer. Data are normalized to a sample randomly chosen from the γ-CD8 treated group (n = 6/group, formula image = p ≤ 0.05).

References

    1. Lechler RI, Sykes M, Thomson AW, Turka LA. Organ transplantation--how much of the promise has been realized? Nat Med. 2005;11(6):605–613. - PubMed
    1. Lindahl KF, Wilson DB. Histocompatibility antigen-activated cytotoxic T lymphocytes. II Estimates of the frequency and specificity of precursors. J Exp Med. 1977;145(3):508–522. - PMC - PubMed
    1. Fishman JA. Infection in solid-organ transplant recipients. N Engl J Med. 2007;357(25):2601–2614. - PubMed
    1. Ajithkumar TV, Parkinson CA, Butler A, Hatcher HM. Management of solid tumours in organ-transplant recipients. Lancet Oncol. 2007;8(10):921–932. - PubMed
    1. Sayegh MH, Remuzzi G. Clinical update: immunosuppression minimisation. Lancet. 2007;369(9574):1676–1678. - PubMed

Publication types