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
. 2017 Sep 20:8:1152.
doi: 10.3389/fimmu.2017.01152. eCollection 2017.

CD28 Blockade Ex Vivo Induces Alloantigen-Specific Immune Tolerance but Preserves T-Cell Pathogen Reactivity

Affiliations

CD28 Blockade Ex Vivo Induces Alloantigen-Specific Immune Tolerance but Preserves T-Cell Pathogen Reactivity

Barbara Dillinger et al. Front Immunol. .

Abstract

Donor T-cells contribute to reconstitution of protective immunity after allogeneic hematopoietic stem cell transplantation (HSCT) but must acquire specific tolerance against recipient alloantigens to avoid life-threatening graft-versus-host disease (GvHD). Systemic immunosuppressive drugs may abrogate severe GvHD, but this also impedes memory responses to invading pathogens. Here, we tested whether ex vivo blockade of CD28 co-stimulation can enable selective T-cell tolerization to alloantigens by facilitating CD80/86-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling. Treatment of human allogeneic dendritic cell/T-cell co-cultures with a human CD28 blocking antibody fragment (α-huCD28) significantly abrogated subsequent allospecific immune responses, seen by decreased T-cell proliferation and of type 1 cytokine (IFN-γ and IL-2) expression. Allo-tolerization persisted after discontinuation of CD28 blockade and secondary alloantigen stimulation, as confirmed by enhanced CTLA-4 and PD-1 immune checkpoint signaling. However, T-cells retained reactivity to pathogens, supported by clonotyping of neo-primed and cross-reactive T-cells specific for Candida albicans or third-party antigens using deep sequencing analysis. In an MHC-mismatched murine model, we tolerized C57BL/6 T-cells by ex vivo exposure to a murine single chain Fv specific for CD28 (α-muCD28). Infusion of these cells, after α-muCD28 washout, into bone marrow-transplanted BALB/c mice caused allo-tolerance and did not induce GvHD-associated hepatic pathology. We conclude that selective CD28 blockade ex vivo can allow the generation of stably allo-tolerized T-cells that in turn do not induce graft-versus-host reactions while maintaining pathogen reactivity. Hence, CD28 co-stimulation blockade of donor T-cells may be a useful therapeutic approach to support the immune system after HSCT.

Keywords: CD28 blockade; alloantigen specific; ex vivo; graft-versus-host disease; preserved pathogen reactivity; tolerance.

PubMed Disclaimer

Figures

Figure 1
Figure 1
Schema of in vitro allo-tolerization and retained pathogen reactivity by α-huCD28-mediated blockade of human T-cells. Alloantigen binding to the respective T-cell receptor (TCR) concurrently with CD28 blockade by α-huCD28 potentially tolerizes human T-cells, while CD80/86 co-stimulatory molecules remain accessible to negative regulators such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) (top). Human T-cells are co-cultured with MHC-mismatched human dendritic cells (DCs) presenting alloantigen (primary mixed leukocyte reaction), in the presence of the CD28 blocker α-huCD28. After 7 days of culture, T-cells are washed, rested for 2 days in the absence of α-huCD28, and re-stimulated with (A) the same alloantigen (fresh allogeneic DCs), (B) Candida albicans (autologous DCs), or (C) third-party alloantigen (third-party DCs).
Figure 2
Figure 2
CD28 blockade mediates persistent reduction of alloresponses. Proliferation of human T-cells co-cultured with allogeneic dendritic cells (DCs) in vitro in a primary or secondary allogeneic mixed leukocyte reaction (MLR). (A) Representative histogram and dotplot analyses of the percentage of carboxyfluorescein succinimidyl ester (CFSE)/CPD dilution and the mean fluorescence intensity of CD3+ T-cells in the primary (upper panel) and secondary MLR (lower panel), with (+) or without (−) α-huCD28. Quadrants were adjusted according to unproliferated (CFSEpos/CPDpos) cells. (B) Proliferation (CFSEneg/CPDneg) of human CD3+ T-cells and FACS-sorted naive (CD45RA+/RO) or memory (CD45RA/RO+) T-cells analyzed by flow cytometry. Proliferation is depicted as number of proliferated cells without (○) or with (▲) α-huCD28 after a 7-day primary MLR (left panel) and a 3-day secondary MLR (right panel). Lines represent results of different biological replicates; points represent the mean of triplicate determinations. Different recipient–donor pairs were used as biological replicates. ***p < 0.001, **p < 0.005, *p < 0.05.
Figure 3
Figure 3
CD28 blockade inhibits Th1 cytokines in secondary allo-mixed leukocyte reactions (MLRs). Th1-specific cytokine expression of FACS-sorted naive and memory T-cells in primary or secondary allo-MLRs. (A,C) Representative dotplots show IL-2 and IFN-γ expression of naive (A) or memory (C) T-cells treated with (+) or without (−) α-huCD28 in a primary and, after discontinuation of CD28 blockade, in a secondary MLR after 24 h. (B,D) Percentage of IFN-γ, IL-2, IFN-γ/IL-2 expressing, and IFN-γ/IL-2 negative naive (B) or memory (D) T-cells, with (▲) or without (○) prior α-huCD28 exposure, in a secondary MLR after 24 h. Pairs of points are the means of triplicate determinations of individual different recipient–donor biological samples. ***p < 0.001, **p < 0.005, *p < 0.05.
Figure 4
Figure 4
CD28 blockade induces checkpoint signaling. Expression of programmed death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) on sorted naive T-cells 24 h after a secondary allogeneic mixed leukocyte reaction (MLR) without (−) or with (+) prior α-huCD28. Cells were gated according to: CD3+/live cells/CD4+/CTLA-4++ or CD3+/live cells/CD4+/PD-1+CTLA-4++. CTLA-4++ and PD-1+CTLA-4++ α-huCD28 pre-treated cells were analyzed for cellular proliferation by carboxyfluorescein succinimidyl ester (CFSE)/CPD dilution. (A) Representative dotplots of PD-1 and CTLA-4 expression on naive CD4+ T-cells in a secondary MLR (day 1). Numbers represent percent within the CD3+ cell population. Cumulative results of CTLA-4 and PD-1 expressions of naive T-cells with (▲) or without (○) prior α-huCD28 after a 1 day secondary allo-MLR. Points represent the mean of replicates from different recipient–donor pairs. (B) T-cells with prior CD28 blockade, highly expressing CTLA-4 or PD-1/CTLA-4, were analyzed for proliferation (CFSEneg/CPDneg, proliferated). Representative dotplots of CFSE/CPD staining are shown; bar graphs show the cumulative results of percent proliferated T-cells. (C) CD3+ T-cell lysates from a primary MLR with (+) or without (−) α-huCD28 for 0–96 h were examined by Western blot to detect phospho-protein phosphatase 2A (pPP2A), total protein phosphatase 2A (PP2A), Akt phosphorylated on threonine 308 (pAkt Thr) or serine 473 residues (pAkt Ser), total Akt, and vinculin as a control. For densitometric analysis (ratio of phosphorylated/total protein) the ImageJ program was used. *p < 0.05.
Figure 5
Figure 5
CD28-tolerized T-cells retain pathogen reactivity. Proliferation of human CD3+ T-cells after re-stimulation with Candida albicans or foreign third-party alloantigen. (A–C) Data points for primary mixed leukocyte reactions (MLRs) are derived from Figure 2B (+ α-huCD28) for comparison. The number of proliferated cells in primary MLRs ranged from 1 × 102 to 9 × 103. T-cell proliferation, shown as number of proliferated cells, was analyzed by flow cytometry after a 7-day re-stimulation with (A) autologous dendritic cells with C. albicans, (B) without C. albicans as a control for possible antigen-independent proliferation, or (C) with foreign third-party alloantigen. Each point is the mean of triplicate determinations of different recipient–donor pairs used as biological replicates. **p < 0.005; ns = not significant, p > 0.05.
Figure 6
Figure 6
Expansion of allo- versus pathogen-reactive T-cell clones after α-huCD28-mediated blockade. Clonal frequency of CD3+ T-cell cultures from a primary recipient–donor pair mixed leukocyte reaction (MLR) and re-stimulation with either 1st party alloantigen, Candida albicans, or third-party alloantigen, as analyzed by deep TCRβ sequencing. (A) Expansion of T-cell clones in a primary MLR without (gray bars) or with α-huCD28 (black bars) and a secondary MLR in the absence of α-huCD28. Top 200 clones are ranked from their lowest to highest frequency in cultures without α-huCD28. The top 10 clones are represented individually in all graphs. (B) Clonal expansion after allo-tolerization and re-stimulation with C. albicans or third-party alloantigen. The frequency of antigen-independent expanded T-cell clones (T-cells stimulated with autologous DCs without C. albicans) was subtracted from the Candida-specific clonal expansion. The top 200 clones were ranked from high to low abundance in C. albicans (red bars) or third-party alloantigen (green bars) re-stimulation cultures and compared to allo-tolerized primary MLRs (black bars). For an in depth investigation, the top 10 most expanded clones are represented individually in all graphs. Clone numbers 2 and 4–10 (left graph) and 2–7 and 9–10 (right graph) are present in primary MLRs, but in low numbers ranging from 0.004 to 0.039%. (C) Unique clones expanded upon stimulation with C. albicans (red bars) or third-party alloantigen (green bars) but not present in secondary MLRs with 1st party alloantigen. Clones are ranked from high-to-low abundance in the top 200 clonal repertoire (numbers at the bottom of the figure).
Figure 7
Figure 7
Ex vivo co-stimulation blocked T-cells do not induce graft-versus-host disease (GvHD) in an MHC-mismatched murine model. Experimental schema (A): T-cells isolated from C56BL/6 spleens were co-cultured with BALB/c dendritic cells (DCs) for 5 days with or without α-muCD28. Lethally irradiated BALB/c mice were transplanted with C56BL/6 bone marrow (BM) and FACS-sorted viable T-cells from mixed leukocyte reaction (MLR) cultures. Mice were monitored for signs of GvHD for 44 days. (B) T-cell proliferation: depicted by the number of proliferated C56BL/6 T-cells upon stimulation with BALB/c DCs together with (+) or without (−) α-muCD28 after a 5-day MLR (left graph), before infusion; cytolytic activity: cytotoxicity of alloreactive CD8+ and CD4+ T-cells (right graph) against MC38 target cells after 21 h is shown. (C) GvHD score of BALB/c mice transplanted with C57BL/6 BM alone (●, n = 6) or together with non-tolerant (freshly isolated) T-cells (■, n = 7), or T-cells without (○, n = 15) or with prior α-muCD28 treatment ex vivo (▲, n = 12). (D) H&E staining of BALB/c livers, necrotic lesions are indicated by black borders, 10× magnification. Necrosis was assessed by quantifying the affected areas using the ImageJ program (− α-muCD28 n = 11, + α-muCD28 n = 11, freshly isolated Tc n = 5, and BM only n = 6). Outliers were removed by applying Grubbs’ test (alpha = 0.1). The sum of the areas of all measurable lesions in a high power field is depicted. Bars represent 100 µm. (E) Dilated blood vessels are indicated by black arrows, 4× magnification. Vasodilation was quantified as the mean of all vessel diameters in a high power field (− α-muCD28 n = 11, + α-muCD28 n = 10, freshly isolated Tc n = 4, BM only n = 4). Outliers were removed by applying Grubbs’ test (alpha = 0.1). Bars represent 500 µm. Image acquisition in (D,E) was done with a Nikon Eclipse 80i microscope with a build on camera Nikon DS-Fi1. Magnification of the object lenses was 500 μm = 4×, 100 μm = 10×. NIS-Elements F4.30.00 software was used for image acquisition. Data shown comprise two separate experiments, scored in a blinded fashion independently by two observers. **p < 0.005, *p < 0.05.

References

    1. Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet (2009) 373(9674):1550–61.10.1016/S0140-6736(09)60237-3 - DOI - PMC - PubMed
    1. Hatzimichael E, Tuthill M. Hematopoietic stem cell transplantation. Stem Cells Cloning (2010) 3:105–17.10.2147/SCCAA.S6815 - DOI - PMC - PubMed
    1. Koyama M, Hill GR. Alloantigen presentation and graft-versus-host disease: fuel for the fire. Blood (2016) 127(24):2963–70.10.1182/blood-2016-02-697250 - DOI - PubMed
    1. Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood (2008) 112(12):4371–83.10.1182/blood-2008-03-077974 - DOI - PubMed
    1. Storek J, Geddes M, Khan F, Huard B, Helg C, Chalandon Y, et al. Reconstitution of the immune system after hematopoietic stem cell transplantation in humans. Semin Immunopathol (2008) 30(4):425–37.10.1007/s00281-008-0132-5 - DOI - PubMed

LinkOut - more resources