Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Common Gamma Chain Cytokines Promote Rapid In Vitro Expansion of Allo-Specific Human CD8+ Suppressor T Cells

  • Yuming Yu,

    Affiliations Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America, Department of Organ Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China

  • Jennifer R. Zitzner,

    Affiliation Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Josetta Houlihan,

    Affiliation Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Nancy Herrera,

    Affiliation Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Luting Xu,

    Affiliation Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Joshua Miller,

    Affiliation Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • James M. Mathew,

    Affiliation Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Anat R. Tambur,

    Affiliation Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Xunrong Luo

    xunrongluo@northwestern.edu

    Affiliations Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

Abstract

Human CD8+ regulatory T cells, particularly the CD8+CD28 T suppressor cells, have emerged as an important modulator of alloimmunity. Understanding the conditions under which these cells are induced and/or expanded would greatly facilitate their application in future clinical trials. In the current study, we develop a novel strategy that combines common gamma chain (γc) cytokines IL-2, IL-7 and IL-15 and donor antigen presenting cells (APCs) to stimulate full HLA-mismatched allogeneic human CD8+ T cells which results in significant expansions of donor-specific CD8+CD28 T suppressor cells in vitro. The expanded CD8+CD28 T cells exhibit increased expressions of CTLA-4, FoxP3, and CD25, while down-regulate expressions of CD56, CD57, CD127, and perforin. Furthermore, these cells suppress proliferation of CD4+ T cells in a contact-dependent and cytokine-independent manner. Interestingly, the specificity of suppression is restricted by the donor HLA class I antigens but promiscuous to HLA class II antigens, providing a potential mechanism for linked suppression. Taken together, our results demonstrate a novel role for common γc cytokines in combination with donor APCs in the expansion of donor-specific CD8+CD28 T suppressor cells, and represent a robust strategy for in vitro generation of such cells for adoptive cellular immunotherapy in transplantation.

Introduction

Allogeneic organ transplantation has emerged as the current best therapeutic option for selected patients with end stage organ failure. Tolerance induction is highly desirable in this population, because long-term immunosuppressive therapy is associated with significant risks for opportunistic infections, degenerative and metabolic diseases, as well as malignancies. Regulatory T cell (Treg) based therapy has emerged as a promising means for immunomodulation to achieve transplant tolerance. Great interest has been focused on CD4+ Tregs [1], and cell therapies using adoptive transfer of in vitro generated CD4+ Treg cells have demonstrated promising efficacy for immunomodulation in animal models of allogeneic transplantation as well as in clinical trials of human allogeneic bone marrow transplantation for the control of graft versus host disease [2], [3], [4]. However, evidence also suggests that CD8+ Tregs may play an important regulatory role in transplant tolerance [5], [6], [7], [8], in addition to possible immunomodulatory roles in autoimmune disorders [9], [10], cancers [11] and aging [12]. Several subsets of CD8+ Tregs have been observed. Natural CD8+ Tregs have been reported to be CD8+CD25+, CD8+CD122+, or CD8+CXCR3+ in different systems [13], [14], [15]. Induced CD8+ Tregs have also been reported to bear various phenotypic characteristics, such as CD28, CD56+, CD57+, CTLA4+, CD103+, CD25+Foxp3+ or LAG3+CCL4+ [5], [16], [17], [18], [19]. These natural or induced CD8+ Tregs exert their suppressive function by cell-cell contact [5] or by producing soluble cytokines [15], [17], and in some circumstances by inducing tolerogenicity in antigen presenting cells (APCs) [20].

Among the various CD8+ Tregs, CD8+CD28 T suppressor cells have emerged as an important modulator of alloimmunity. Human allo-specific CD8+CD28 cells have been shown to emerge after repeated stimulation with allogeneic APCs with supplemental recombinant human IL-2 in vitro [5]. These cells have subsequently been termed “Ts cells”. Similarly, autoantigen-specific CD8+ Tregs including CD8+CD28 cells have been reported to be induced in vitro via stimulation of peripheral T cells obtained from patients with systemic lupus erythematosus (SLE) in the presence of a combination of common gamma chain (γc) cytokines IL-2, IL-7 and IL-15 [21]. These conditions under which CD8+CD28 cells are induced are distinct from those in which CD4+ Tregs are induced, in that the latter frequently require the presence of tolerance promoting agents such as TGF-β, IL-10, or rapamycin in the culture [22], [23], [24], [25]. While the mechanisms by which CD8+CD28 cells are induced under such apparently non-tolerance promoting conditions remain unclear, the induced CD8+CD28 cells nevertheless exhibit potent antigen-specific suppressive capacities [5], [21]. The γc cytokines have also been reported to induce stable loss of CD28 expression in actively dividing CD8+CD28+ T cells [26], [27], thereby promoting the generation of a preponderance of CD8+CD28 T cells. We therefore questioned if γc cytokines in combination with donor APCs may be used to expand donor-specific CD8+CD28 T suppressor cells in large numbers in vitro, and if so, what the mechanisms of donor-specific suppression are.

In this report, we utilized APCs and T cells variably matched for human leukocyte antigen (HLA) class I or class II to study the ability and mechanisms of suppression of the CD8+CD28 T cells generated in vitro by this novel approach of combining allogeneic APCs plus γc cytokines. Our findings indicate that large numbers of CD8+CD28 T cells with potent donor-specific suppressive capacity can be effectively generated in vitro using this approach. Furthermore, their suppressive capacity is contact-dependent and is restricted only by the donor HLA class I antigens but promiscuous to HLA class II antigens. To our knowledge, this is the first report demonstrating a role of combination γc cytokines in the in vitro generation of large numbers of human donor-specific CD8+CD28 suppressor cells. Findings from this study may have important implications for designing future Treg based therapies for prevention of rejection in human organ or tissue transplantation.

Results

Donor APCs plus γc cytokines induce rapid expansion of CD8+CD28 T cells in culture

The CD28 cell population accounts for a small fraction of freshly isolated human CD8+ T cells from PBMCs (5.5%, Figure 1A, top panel, Day 0). Fresh CD8+ T cells were stimulated with HLA-A, -B and -DR complete-mismatched APCs in the presence of IL-2 alone or a combination of γc cytokines IL-2, IL-7 and IL-15 as described in Materials and Methods [21]. As shown in Figure 1A bottom panels (Day 6, Day 9), culture in the presence of IL-2 alone allowed only minimal increase of the CD28 population over time. In contrast, culture in the presence of a combination of γc cytokines IL-2, IL-7 and IL-15 significantly increased this population from 5.5% of all CD8+ cells on day 0 to 45.7% (51.5+/−8.5%) on day 6, and 55.5% (58.5+/−7.2%) on day 9 (Figure 1A), likely representing down-regulation of cell surface CD28 during the in vitro cell activation as previously described [26], [27]. The total number of CD8+CD28 recovered at the end of the 9 day culture increased by ∼26.7+/−7.5 fold (Figure 1B). The most dramatic increase of this population was observed when IL-15 was added to the culture, with moderate effect observed with IL-7 on the expansion of this population (data not shown).

thumbnail
Figure 1. Donor APCs plus γc cytokines induce rapid expansion of CD8+CD28 T cells in culture.

Freshly purified CD8+ T cells (2×106 per well) from healthy volunteers were stimulated by HLA-A, B and DR mismatched allogeneic APCs (1×106 per well) for 9 days in 24-well plates supplemented with IL-2 alone or a combination of IL-2, IL-7 and IL-15. A, Expression of CD28 on CD8+ cells over time. Histograms were gated on the CD8+ population. B, Expansion of cell numbers of the CD8+CD28 population over time by donor APC plus γc cytokines stimulation. The numbers of the CD8+CD28 cells per well of 24-well culture plates were calculated for day 0, 6 and 9. Data shown in A are representative of three independent experiments. Data shown in B is the average of three independent experiments.

https://doi.org/10.1371/journal.pone.0028948.g001

In vitro expanded CD8+CD28 T cells suppress CD4+ T cells proliferation in a donor-specific manner

CD8+CD28 T cells were generated from individual A by culturing with allogeneic APCs from individual B (designated as B-APC) in the presence of the γc cytokine combination as described above. After 9 days of culture, the resulting CD8+CD28 T cells were isolated (purity>95%) and used as suppressors (S) to test for their suppressive capacity in mixed lymphocyte reactions (MLRs) using CD4+ T cells from individual A (designated as A-T4) as responders (R). Suppressor to responder ratios (S∶R) of 0.5∶1, 0.1∶1 and 0.02∶1 were tested (with the cell number of “R” kept constant). As shown in Figure 2A, when APCs from the original priming stimulator (B-APC) were used as stimulators of the MLRs, proliferation of A-T4 (measured by CFSE dilution) was markedly suppressed in a dose-dependent manner by the in vitro expanded CD8+CD28 T cells. Suppression was also confirmed by using 3H-thymidine incorporation as a read-out (Figure 2B). In contrast, when APCs from an HLA-A, -B and -DR mismatched indifferent stimulator (I-APC) were used as stimulators of the MLRs, CD8+CD28 cells exhibited only marginal suppression at the highest S∶R ratio tested (0.5∶1, shown in Fig. 2A, middle panels). No suppression could be demonstrated when anti-CD3/CD28 coated Dynabeads were used for stimulating the CD4+ T cells (Figure 2A, bottom panels). Taken together, our results indicate that the CD8+CD28 T cell generated from donor APCs plus γc cytokine-driven in vitro expansion suppress CD4+ T cells proliferation only in a donor-specific manner.

thumbnail
Figure 2. In vitro expanded CD8+CD28 T cells suppress CD4+ T cells proliferation in a donor-specific manner.

CD8+ T cells from donor A were stimulated with APCs from donor B plus γc cytokines for 9 days, followed by enrichment of the CD8+CD28 population as described in Materials and Methods. 5×104 CFSE labeled purified responder CD4+ T cells (R) from donor A were stimulated with 5×104 APCs from donor B (B-APC) or from an indifferent donor (I-APC, HLA-A, B and DR fully mismatched with donor B), or 1×104 anti-CD3/CD28 coated Dynabeads in triplicates in 96-well plates. The in vitro expanded CD8+CD28 T cells were added as putative suppressors (S) at S∶R ratios of 0.5∶1, 0.1∶1 and 0.02∶1 (with the cell number of “R” kept constant). Proliferation of the CD4+ cells was measured by CFSE dilution (A) or by 3H thymidine uptake (B). Data shown are representative of three independent experiments. * P = 0.002; ** P = 0.009; *** P = 0.008.

https://doi.org/10.1371/journal.pone.0028948.g002

Suppression by the in vitro expanded CD8+CD28 T cells is restricted by donor HLA class I antigens but promiscuous to HLA class II antigens

To determine whether the donor specificity observed in the above suppression of proliferation was restricted via recognition of HLA class I antigens only, we examined the ability of the CD8+CD28 T cells generated from culturing with B-APC to suppress proliferation stimulated by APCs from a third-party donor (C-APC) who shared identical HLA class I but not class II antigens with individual B. The ability of suppressing C-APC driven proliferation was compared with that of suppressing I-APC driven proliferation (I-APC were from an indifferent individual who was mismatched with individual A for both HLA class I and class II antigens). Table 1 provides one example of the HLA class I and class II antigen typing of individuals A, B, C, and I used for one of three such experiments. As shown representatively in Figure 3, proliferation of A-T4 was profoundly suppressed when B-APC was used as the stimulator as expected. Interestingly, a significant suppression was also observed, albeit at a slightly reduced level, when C-APC was used as the stimulator. In contrast, suppression was almost completely lost when I-APC was used as the stimulator. These data support the hypothesis that suppression by the in vitro expanded CD8+CD28 T cells is mediated via specific interactions between the T cell receptor (TCR) on the CD8+ cells and donor HLA class I molecules on the APCs, and such interactions can lead to suppression of CD4+ cells with TCRs specific for HLA class II molecules co-expressed on the same interacting APCs (the C-APC in this case) but different from those expressed on the original priming APCs (the B-APC in this case). This finding corroborates with the previous observation that CD8 suppressor cells are capable of exerting linked suppression via interactions with APCs [28].

thumbnail
Figure 3. Suppression by the in vitro expanded CD8+CD28 T cells is restricted by donor HLA class I antigens.

Suppression assays were set up as described in Figure 2, with the exception that the stimulator APCs for CD4+ proliferation were from either the priming donor (B-APC), an indifferent donor (I-APC, HLA-A, B and DR fully mismatched with donor B), or a partially matched donor (C-APC, who shared identical HLA class I, but not class II antigens, with donor B). Proliferation of the CD4+ cells was measured by CFSE dilution. Data shown are representative of three independent experiments using three pairs of donors B, C and I. Table 1 provides one example of the HLA class I and class II antigen typing of individuals B, C, and I used for one of three such experiments.

https://doi.org/10.1371/journal.pone.0028948.g003

thumbnail
Table 1. Representative HLA typing of donors used for experiments demonstrating HLA class I antigen specificity shown in Figure 3.

https://doi.org/10.1371/journal.pone.0028948.t001

Suppression by the in vitro expanded CD8+CD28 T cells is contact dependent but IFN-γ or TGF-β independent

The role of cell-cell contact in suppression was examined using transwell assays as shown in Figure 4A. The lower chamber was plated with CFSE-labeled responder cells (R) and stimulator cells (B-APC), and the in vitro expanded CD8+CD28 T cells (S) were added either in the lower chamber to allow cell-cell contact or in the upper chamber to prevent cell-cell contact. As shown in Figure 4A, suppression of proliferation by CD8+CD28 T cells was abolished when these cells were plated in the upper chamber of the transwells, indicating that suppression by these cells requires cell-cell contact between the CD8+CD28 T cells, stimulator APCs and/or the responder CD4+ T cells. This finding is consistent with the characteristic donor HLA class I restriction of the suppressive activity of the in vitro expanded CD8+CD28 T cells observed in Figure 3.

thumbnail
Figure 4. Suppression by the in vitro expanded CD8+CD28 T cells is contact dependent but IFN-γ or TGF-β independent.

Suppression assays were set up as described in Figure 2 at an S∶R ratio of 0.5∶1. Proliferation of the CD4+ cells was measured by CFSE dilution. A, Transwell assays: the lower chambers of 24-well transwell plates were plated with CFSE-labeled responder cells (3×105 of R) and stimulator cells (3×105 of B-APC), and the in vitro expanded CD8+CD28 T cells were added either in the lower chamber to allow cell-cell contact or in the upper chamber to prevent cell-cell contact. Data shown are representative of three independent experiments. B, Control suppression without any blocking antibodies. C, Suppression in the presence of anti-human IFN-γ antibody or its isotype control antibody at concentrations of 1 µg/ml, 10 µg/ml and 100 µg/ml. D, Suppression in the presence of anti-human TGF-β antibody or its isotype control antibody at concentrations of 1 µg/ml, 10 µg/ml and 100 µg/ml. Data shown for B and C are representative of three independent experiments. Abbreviations used for AD: “R”: CD4+ responder cells from donor A; “B-APC”: APC stimulators from donor B; “S”: the in vitro expanded CD8+CD28 T cells.

https://doi.org/10.1371/journal.pone.0028948.g004

Several reports have implicated TGF-β [13], [29], [30] and IFN-γ [8], [31], [32] in the mechanisms of regulation by CD8+ suppressor cells. To test for a potential role of TGF-β or IFN-γ in the suppressive capacity of the in vitro expanded CD8+CD28 T cells in our system, anti-human IFN-γ or anti-human TGF-β antibody was added to the suppression assays described above (baseline suppression without the blocking antibodies is shown in Figure 4B). As shown in Figure 4C and Figure 4D, neither anti-IFN-γ nor anti-TGF-β antibodies at increasing concentrations affected suppression by CD8+CD28 T cells.

In vitro expanded CD8+CD28 T cells do not exhibit cytotoxicity

To test whether cytotoxicity towards the stimulating APCs contributes to the suppression of donor-specific proliferation observed above, we assessed the cytotoxicity of CD8+CD28 T cells using a CFSE-based assay [33]. We designed this assay to specifically determine the ability of the CD8+CD28 T cells to lyse allogeneic donor APCs used as stimulators in the above MLRs. In this assay, “effector cells” were the CD8+CD28 T cells generated by the donor APC plus γc cytokine-driven in vitro cultures as above. The CD8+CD28+ T cells generated by the same cultures were used as control effector cells because of their known cytotoxicity [5], [34], [35]. Target cells were either the original priming APCs which were labeled with a high concentration of CFSE (B-APC-CFSEhigh), or control APCs from an HLA-A, -B, and -DR mismatched indifferent individual which were labeled with a low concentration of CFSE (I-APC-CFSElow). A 1∶1 mixture of I-APC-CFSElow and B-APC-CFSEhigh cells were plated either by themselves or with the same number of the putative (CD8+CD28) or control (CD8+CD28+) effector cells. At 24, 72, and 120 hr of culture, cells were collected and analyzed by FACS to enumerate CFSElow and CFSEhigh cells. The ratio of cell numbers of I-APC-CFSElow over B-APC-CFSEhigh (shown as “R” in Figure 5) over time provided a measure of specific cytolytic effect of the putative effector cells on these two target populations. As shown in Figure 5 left panels, minimal spontaneous lysis was observed as the ratio R remained stable over time in the absence of any effector cells. When the CD8+CD28+ T cells were added as effector cells (Figure 5, middle panels), R increased to 1.5 at 72 hrs and 2.9 at 120 hrs, demonstrating that B-APC-CFSEhigh cells were specifically killed by the CD8+CD28+ T cells. In contrast, when the CD8+CD28 T cells were added as effector cells, R remained unchanged over time, indicating that the CD8+CD28 T cells exhibit no specific cytolysis towards the B-APC cells (Figure 5, right panels).

thumbnail
Figure 5. Cytotoxicity does not contribute to the suppression of proliferation by the in vitro expanded CD8+CD28 T cells.

A CFSE based cytotoxicity assay was set up as follows: target cells were the original priming APCs (for generating the CD8+CD28 T cells) labeled with a high concentration (2.0 µM) of CFSE (B-APC-CFSEhigh), or APCs from an HLA-A, -B, and -DR mismatched indifferent individual labeled with a low concentration (0.2 µM) of CFSE (I-APC-CFSElow). A 1∶1 mixture of I-APC-CFSElow and B-APC-CFSEhigh cells were plated either by themselves (left panels), or with the same number of control (CD8+CD28+, middle panels) or putative (CD8+CD28, right panels) effector cells in 96 U-bottom plates in triplicates. At 24, 72, and 120 hr of culture, cells were collected and analyzed by FACS to enumerate CFSElow and CFSEhigh cells. The ratio of cell numbers of I-APC-CFSElow over B-APC-CFSEhigh was calculated as “R” for all time points. An increase in the value of R indicates specific killing of B-APC-CFSEhigh cells by the effector cells, whereas a stable value of R at 1.0 indicates no specific killing of B-APC-CFSEhigh cells by the effector cells. Data shown are representative of two independent experiments.

https://doi.org/10.1371/journal.pone.0028948.g005

Phenotypic characteristics of the in vitro expanded CD8+CD28 T cells

Phenotypic characteristics of CD8+CD28 T cells were examined before and after donor APCs plus γc cytokine-driven in vitro expansion to determine if reported markers of CD8 Tregs such as CTLA-4, FoxP3, CD25, CD56 and CD57 [5], [16], [17], [18], [19] were also expressed by the CD8+CD28 T cells generated by our protocol. As shown in Figure 6A, among freshly isolated CD8+ T cells prior to in vitro stimulation, CD8+CD28 T cells comprised a small fraction. At baseline, they expressed CD56 (35.0%), CD57 (76.5%), CD62L (28.0%), CD127 (13.6%), and perforin (36.6%), but exhibited minimal expressions of CTLA-4, FoxP3, CD25 (Figure 6A). Following donor APCs plus γc cytokine-driven expansion, CD28 cells became the predominant population among CD8+ cells, and they significantly up-regulated expressions of CTLA-4 (78.7%), FoxP3 (16.0%), and CD25 (37.5%) compared with basline, while down-regulated expressions of CD56, CD57, CD62L, CD127, and perforin (Figure 6B). While the exact percentages of these markers varied between the CD8+CD28 cells from different individuals (data not shown), the trend of up- or down-regulations before and after stimulation in cultures remained the same (1 of 3 representative experiments is shown in Figure 6).

thumbnail
Figure 6. Phenotypic characteristics of the in vitro expanded CD8+CD28 T cells.

CD8+ T cells freshly isolated from healthy volunteers (A) or after in vitro stimulation with donor APCs plus γc cytokines for 9 days (B) were analyzed for cell surface or intracellular markers by multichromatic flow cytometry. Date shown are representative of three independent experiments.

https://doi.org/10.1371/journal.pone.0028948.g006

Discussion

In this study, we reported the novel finding that large numbers of donor-specific human CD8+CD28 T suppressor cells can be generated in vitro using a combination of donor APCs and γc cytokines stimulated proliferation. Common γc cytokines IL-2, IL-7 and IL-15 play critical roles in T cell homeostasis [36]. Recent reports have demonstrated that IL-15 can promote the generation and proliferation of CD8+ memory T cells when stimulated by anti-CD3 and anti-CD28 antibodies [12], [26]. In contrast, a combination of γc cytokines including IL-2, IL-7 and IL-15 has been used to expand CD8+Foxp3+ Treg in vitro from autoimmune patients with SLE who have undergone autologous bone marrow transplant [21]. Human allo-specific CD8+CD28 suppressor T cells have been reported to be generated by two rounds of stimulation by allogeneic APCs in which IL-2 was the only cytokine added during the last four days of cultures [20]. These cells have been termed as Ts cells. Here, we demonstrated that the combination of IL-2, IL-7 and IL-15, compared with IL-2 alone, was superior in inducing significant expansions of allo-specific CD8+CD28 cells in cultures activated by donor APC stimulation (Figure 1A, 1B), and that the expanded CD8+CD28 cells were highly functionally suppressive. The role of each individual cytokine in the expansion versus inducing the “suppressive” characteristics of this cell population remains to be further elucidated.

The phenotypic and functional characteristics of the CD8+CD28 T cells generated in our protocol with a combination of γc cytokines closely resemble those of previously described Ts cells [19], [20]. They both up-regulate CTLA-4, FoxP3 and CD25, and have variable degrees of down-regulation of CD62L during expansion; possess minimal cytotoxicity; and exhibit suppressive capacity in a contact dependent fashion. We further demonstrated that our in vitro expanded CD8+CD28 T cells exert linked suppression of CD4 T cells of a different HLA class II specificity as long as the HLA class II molecules are co-expressed with the priming HLA class I molecules on the same APCs. This has also been previously indirectly demonstrated as a characteristic of Ts cells [5]. The ability to generate large numbers of functionally suppressive donor-specific CD8+CD28 T cells that closely mimic Ts cells with this novel approach of using common γc cytokines thus has significant potential for facilitating future clinical use of Treg cell-based immunotherapy for tolerance induction in transplantation.

Several mechanisms of suppression by CD8 suppressor cells have been described. These include direct cytotoxicity [37], secretion of inhibitory cytokines [32], [38], down-regulation of co-stimulatory molecules [5], [19], and generation of tolerogenic APCs [20]. In our suppression assays, the CD8+CD28 T cells generated by donor APCs plus γc cytokines suppress proliferation of CD4+ T cells in a donor-specific and dose-dependent manner. Interestingly, distinct from CD4 Treg cells, the CD8+CD28 T cells do not have the ability to suppress proliferation driven by non-specific stimulation by anti-CD3 and anti-CD28 coated Dynabeads. This finding indicates the importance of the interacting APCs in mediating the suppression by the CD8+CD28 T cells. Further studies characterizing phenotypic and functional changes of the interacting APCs will provide definitive evidence for this hypothesis. Furthermore, the transwell assay showed that separation of the CD8+CD28 T cells alone or CD8+CD28 T cells together with stimulator APCs from the responder CD4+ T cells abolished the ability of the CD8+CD28 T cells to suppress. These data further indicate that suppression of CD8+CD28 T cells is mediated through cell-cell contact in a three-cell system, i.e. not only between the CD8+CD28 T cells and the APCs, but also between the APCs (or the CD8+CD28 T cells) and the responding CD4+ T cells.

The finding that suppression by the CD8+CD28 T cells is restricted by HLA class I but is promiscuous towards HLA class II specificities with which the responding CD4 TCRs interact suggests a potential mechanism for linked suppression. Linked suppression classically depicts a situation in which co-expression of an indifferent antigen and a tolerant antigen on the same graft allows acceptance of the indifferent antigen in a host previously tolerized to the tolerant antigen [39]. Dendritic cells are thought to play a critical role in the process of linked suppression [39]. In our studies, C-APC cells co-express both matched HLA class I antigens with the priming donor B-APC and mismatched HLA class II antigens with the priming donor B-APC. Therefore linked suppression by the CD8+CD28 T cells occurs via C-APC cells, and proliferation of CD4+ responder to the mismatched class II antigens is suppressed. These findings have significant implications for designing and understanding the therapeutic value of this CD8+ suppressor population.

The role of cytotoxicity in mechanisms of regulation by CD8+ suppressor cells has been debated. Suppressive CD8+CD28 Ts cells generated by repeated stimulations with alloantigens have been shown to have no cytotoxicity towards the stimulating allogeneic APCs [5]. In contrast, Zheng et al [37] reported that human CD8high T cells induced by allogeneic CD40-activated B cells have regulatory property that is at least in part mediated through cytotoxicity towards the allogeneic priming PBMC. In our in vitro CFSE-based cytotoxicity assay (Figure 5), it appears that the CD8+CD28 T cells generated by our protocol do not possess strong cytotoxicity towards their priming APCs. This is consistent with the down-regulation of perforin in these cells after culturing (Figure 6). It should be noted, however, that cytotoxicity and other possible regulatory mechanisms, such as cytokine production or those mediated through high levels of expression of FoxP3 and CTLA-4, need not be mutually exclusive.

Both cytokine TGF-β [13], [29], [30] and IFN-γ [8], [31], [32] have been implicated in the regulatory mechanisms of CD8+ suppressor cells in different studies. In our system, neither anti-IFN-γ nor anti-TGF-β antibody reversed the suppressive effect of the expanded CD8+CD28 T cells (Figure 4C, D). CD56 and CD57 are two markers of natural killer cells previously reported to over-express on CD8+CD28 Ts cells [19]. However, in our culture system the expression of these markers on the CD8+CD28 cells decreased after culture, suggesting that they may not play a major role in the functionality of expanded CD8+CD28 cells in our system.

Two proteins, FoxP3 and CTLA-4, are discernibly up-regulated in the expanded CD8+CD28 T cells. FoxP3 has been described as an important marker for CD8+ suppressor cells in humans [18], [20], [40], [41]. However, unlike in mice, in humans non-regulatory T cells including effector cells can also up-regulate FoxP3 expression transiently upon activation [42], [43]. Moreover, there are also CD8+ suppressor cells that are not characterized with FoxP3 expression [16], [32], [38]. Whether or not FoxP3 up-regulation in our in vitro expanded CD8+CD28 T cells contribute to their suppressive function remains to be elucidated. CTLA-4 is another marker described in CD8+ suppressor cells [18], [20], [40], [41]. CTLA-4 is a homologue of CD28, and is known as a negative co-stimulatory receptor which upon signaling from CD80 and CD86 delivers inhibitory signals to T cells to down-regulate T cell activation [44], [45]. Recent data also suggest that signaling between CTLA-4 and CD80/CD86 can be bi-directional. CTLA-4 expressed on surface of CD4+ Tregs has been shown to interact with CD80/CD86 expressed on APCs and subsequently down-regulate their expression, possibly via a process of trans-endocytosis [46], thereby decreasing the ability of APCs to stimulate T cells through CD28 [47], [48]. CTLA-4 has also been reported to up-regulate indolamine 2,3 dioxygenase (IDO) in some subsets of APCs as yet another mechanism for regulating T cell responses [49]. Given the profound up-regulation of this molecule (from 0.2% to 78.7%, Figure 6) and simultaneous down-regulation of CD28 in our CD8+CD28 T cells, CTLA-4 mediated negative signaling to the interacting APCs is a highly attractive potential mechanism of suppression by this cell population.

While robust suppression is observed from our in vitro expanded CD8+CD28 T cells, an important issue which remains to be resolved is how these cells would behave in vivo upon adoptive transfer. Significant plasticity has been described of CD4+CD25+FoxP3+ T cells. Studies of in vivo viability, stability and functionality of the expanded human CD8+CD28 T cells in our protocol will necessitate the use of functional humanized mouse models of allogeneic immune responses. Such studies are critical before this approach can be safely translated into clinical applications and are currently being actively pursued in our laboratory.

In conclusion, our findings show that donor APCs plus γc cytokines can be harnessed for efficient in vitro expansion of human donor-specific CD8+CD28 T suppressor cells in a simple and robust culture system. This approach may hold promise for clinical application of using regulatory T cell-based immunotherapy for tolerance induction in transplantation in the future.

Materials and Methods

Ethics Statement

This study was approved by the Northwestern Institutional Review Board and written consent was obtained from the participants.

Human subjects and HLA typing

Peripheral blood mononuclear cells (PBMCs) were obtained from healthy volunteers. HLA typing was performed by the Northwestern Histocompatibility Laboratory using molecular methods. Donors for T cells and APCs were selected for their HLA- A, B and DR compatibility or incompatibility based on the specific requirements of individual experiments.

Isolation of PBMCs and cell subsets

PBMCs were isolated from fresh whole blood using Lymphocyte Separation Medium (Mediatech Inc., Manassas, VA) by density gradient centrifugation. CD8+ and CD4+ T cells were purified from PBMCs using CD8 or CD4 isolation kits (Miltenyi Biotec) through positive selection according to the manufacturer's protocols. Purities for both T cells were confirmed by flow cytometry and were routinely >98%. APCs were isolated from PBMCs by depletion of CD3+ cells using CD3 microbeads (Miltenyi Biotech). After culturing of the CD8+ cells for indicated number of days, CD28+ cells were removed by positive selection using human CD28 MicroBead Kit (Miltenyi Biotech, purity>99%), and the flow-through was collected as CD28 cells (purity>95%). Again, purity of all isolated cells was confirmed by flow cytometry.

In vitro generation and expansion of CD8+CD28 cells with allogeneic APCs and γc cytokines

2×106 purified CD8+ T cells from individual A were cultured with 1×106 HLA-A, -B, -DR mismatched APCs from individual B in 2 ml culture medium (RPMI-1640 supplemented with 15% normal human blood group AB serum, 2 mM L-glutamine, 10 mM HEPES and 1×antibiotic-antimycotic solution; all from Gibco-BRL, Gaithersburg, MD) supplemented with IL-2 (20 U/ml), IL-7 (50 ng/ml) and IL-15 (50 ng/ml) (PeproTech Inc., Rocky Hill, NJ) in 24-well plates at 37°C in 5% CO2. Supplemented culture medium was changed on days 4, 7 and 8 (by replacing 1 ml of the culture medium with fresh medium containing cytokines). Cells in each well were split into two wells on day 6 and harvested on day 9, and the CD28 population was isolated as described above.

Suppression of donor-specific proliferation by in vitro generated CD8+CD28 cells

5×104 CFSE labeled purified CD4+ T cells from individual A (A-T4) were used as “responders (R)” and stimulated with 5×104 APCs from the original priming donor (individual B; B-APC). APCs from an HLA-A,-B,-DR complete-mismatched indifferent donor (I-APC) or 1×104 anti-CD3/anti-CD28 Dynabeads (Invitrogen, Carlsbad, CA) were used as third party or non-specific stimulation controls respectively. All cultures were prepared in triplicates and incubated in 96-well U-bottom plates in 37°C 5% CO2 incubator. The CD8+CD28 cells were added as “suppressor (S)” at S∶R ratios of 0.5∶1, 0.1∶1 and 0.02∶1 (the cell number of “R” was kept constant). CFSE dilution was assessed on day 7 to determine extent of proliferation. In selected experiments, anti-human IFN-γ (eBioscience Inc.), anti-human TGF-β antibody (BioXcell, West Lebanon, NH), or respective isotype controls were added to cultures at concentrations of 1 µg/ml, 10 µg/ml or 100 µg/ml. Parallel cultures were analyzed using 3H-thymidine incorporation. 1 µCi 3H-thymidine (PerkinElmer, San Jose, CA) was added per well for the last 18 hrs of a 7-day culture, and incorporated radioactivity was measured as counts per minute (CPM) with a Perkin-Elmer scintillation counter.

Transwell experiments

The lower chambers of 24-well transwell plates were plated with either 3×105 CFSE labeled naïve CD4+ T cells from individual A (A-T4), or with A-T4 and 3×105 priming APCs from individual B (B-APC) in the presence or absence of 3×104 CD8+CD28 cells (total volume 600 µl). The upper chambers were plated with medium only, CD8+CD28 cells only, or CD8+CD28 cells plus priming APCs (B-APC). Cells collected from the lower chamber after 7 days of culture were assessed by FACS for CFSE dilution.

Cytotoxicity by CD8+CD28 cells

CFSE-based cytotoxic assay was set up according to published methods [33] as follows. APCs serving as target cells were labeled with two concentrations of CFSE: high concentration (2.0 µM) for APCs from the priming donor (B-APC-CFSEhigh) and low concentration (0.2 µM) for APCs from an HLA-A,-B,-DR complete-mismatched indifferent donor (I-APC-CFSElow). The in vitro generated CD8+CD28 cells or their CD8+CD28+ counterpart were used as putative or control effector cells respectively. 5×104 effector cells were cultured with 5×104 each of B-APC-CFSEhigh and I-APC-CFSElow together in 96-well U-bottom plates in triplicate wells. Cultured B-APC-CFSEhigh and I-APC-CFSElow in the absence of effector cells were used as controls for spontaneous cytolysis. B-APC-CFSEhigh and I-APC-CFSElow were enumerated by FACS after 0, 24, 72 and 120 hrs of culture. The ratios of cell numbers of I-APC-CFSElow over B-APC-CFSEhigh in the absence or the presence of effector cells were calculated over time to estimate specific killing of B-APC-CFSEhigh.

Multi-color flow cytometric analysis

The following anti-human antibodies and their isotype controls were used: FITC conjugated anti-CD3 (OKT3), CD4(OKT4), CD8(SK1), CD56(MEM188), CD57(TBO1), CD62L(DREG56), CD127(RDR5), Perforin(dG9), all from eBioscience; FITC conjugated anti-human CTLA-4(A3.4H2.H12, Cedarlane USA, Burlington, NC), CD25(BC96), Granzyme B(GB11, Biolegend, San Diego, CA); PE conjugated anti-human Foxp3(eBioscience, San Diego, CA); ECD conjugated anti-human CD8(SFCI21Thy2D3), PE-cyanin 5 (PC5) conjugated anti-human CD28(28.2), PE-cyanin 7 (PC7) conjugated anti-human CD8(SFCI21Thy2D3, Beckman Coulter, Miami, FL). Surface and intra-cellular staining was performed following manufacturers' recommended protocols. Data was acquired on a 5-color FC500 flow cytometer and analyzed using the CXP program (Beckman-Coulter).

Statistical analysis

Comparisons were made by paired Student's t test. Differences were considered significant if the P value was <0.05.

Acknowledgments

We wish to thank Dr. Syamal Datta for helpful discussions and critical reading of this manuscript. We also wish to thank Ms. Sam Malladi for meticulous proof-reading of the manuscript.

Author Contributions

Conceived and designed the experiments: YY ART XL. Performed the experiments: YY JRZ JH NH. Analyzed the data: YY JRZ ART XL. Contributed reagents/materials/analysis tools: LX JM JMM ART. Wrote the paper: YY JRZ ART XL.

References

  1. 1. Sakaguchi S (2005) Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 6: 345–352.
  2. 2. Tsang JY, Tanriver Y, Jiang S, Leung E, Ratnasothy K, et al. (2009) Indefinite mouse heart allograft survival in recipient treated with CD4(+)CD25(+) regulatory T cells with indirect allospecificity and short term immunosuppression. Transpl Immunol 21: 203–209.
  3. 3. Joffre O, Santolaria T, Calise D, Al Saati T, Hudrisier D, et al. (2008) Prevention of acute and chronic allograft rejection with CD4+CD25+Foxp3+ regulatory T lymphocytes. Nat Med 14: 88–92.
  4. 4. Trzonkowski P, Bieniaszewska M, Juscinska J, Dobyszuk A, Krzystyniak A, et al. (2009) First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127− T regulatory cells. Clin Immunol 133: 22–26.
  5. 5. Liu Z, Tugulea S, Cortesini R, Suciu-Foca N (1998) Specific suppression of T helper alloreactivity by allo-MHC class I-restricted CD8+CD28− T cells. Int Immunol 10: 775–783.
  6. 6. Dijke IE, Caliskan K, Klepper M, de Kuiper R, Balk AH, et al. (2009) Donor-specific immune regulation by CD8 lymphocytes expanded from rejecting human cardiac allografts. Am J Transplant 9: 397–403.
  7. 7. Li XL, Menoret S, Bezie S, Caron L, Chabannes D, et al. (2010) Mechanism and localization of CD8 regulatory T cells in a heart transplant model of tolerance. J Immunol 185: 823–833.
  8. 8. Guillonneau C, Hill M, Hubert FX, Chiffoleau E, Herve C, et al. (2007) CD40Ig treatment results in allograft acceptance mediated by CD8CD45RC T cells, IFN-gamma, and indoleamine 2,3-dioxygenase. J Clin Invest 117: 1096–1106.
  9. 9. Konya C, Goronzy JJ, Weyand CM (2009) Treating autoimmune disease by targeting CD8(+) T suppressor cells. Expert Opin Biol Ther 9: 951–965.
  10. 10. Tsai S, Shameli A, Yamanouchi J, Clemente-Casares X, Wang J, et al. (2010) Reversal of autoimmunity by boosting memory-like autoregulatory T cells. Immunity 32: 568–580.
  11. 11. Jarnicki AG, Lysaght J, Todryk S, Mills KH (2006) Suppression of antitumor immunity by IL-10 and TGF-beta-producing T cells infiltrating the growing tumor: influence of tumor environment on the induction of CD4+ and CD8+ regulatory T cells. J Immunol 177: 896–904.
  12. 12. Godlove J, Chiu WK, Weng NP (2007) Gene expression and generation of CD28−CD8 T cells mediated by interleukin 15. Exp Gerontol 42: 412–415.
  13. 13. Cosmi L, Liotta F, Lazzeri E, Francalanci M, Angeli R, et al. (2003) Human CD8+CD25+ thymocytes share phenotypic and functional features with CD4+CD25+ regulatory thymocytes. Blood 102: 4107–4114.
  14. 14. Rifa'i M, Kawamoto Y, Nakashima I, Suzuki H (2004) Essential roles of CD8+CD122+ regulatory T cells in the maintenance of T cell homeostasis. J Exp Med 200: 1123–1134.
  15. 15. Shi Z, Okuno Y, Rifa'i M, Endharti AT, Akane K, et al. (2009) Human CD8+CXCR3+ T cells have the same function as murine CD8+CD122+ Treg. Eur J Immunol 39: 2106–2119.
  16. 16. Uss E, Rowshani AT, Hooibrink B, Lardy NM, van Lier RA, et al. (2006) CD103 is a marker for alloantigen-induced regulatory CD8+ T cells. J Immunol 177: 2775–2783.
  17. 17. Joosten SA, van Meijgaarden KE, Savage ND, de Boer T, Triebel F, et al. (2007) Identification of a human CD8+ regulatory T cell subset that mediates suppression through the chemokine CC chemokine ligand 4. Proc Natl Acad Sci U S A 104: 8029–8034.
  18. 18. Mahic M, Henjum K, Yaqub S, Bjornbeth BA, Torgersen KM, et al. (2008) Generation of highly suppressive adaptive CD8(+)CD25(+)FOXP3(+) regulatory T cells by continuous antigen stimulation. Eur J Immunol 38: 640–646.
  19. 19. Davila E, Kang YM, Park YW, Sawai H, He X, et al. (2005) Cell-based immunotherapy with suppressor CD8+ T cells in rheumatoid arthritis. J Immunol 174: 7292–7301.
  20. 20. Chang CC, Ciubotariu R, Manavalan JS, Yuan J, Colovai AI, et al. (2002) Tolerization of dendritic cells by T(S) cells: the crucial role of inhibitory receptors ILT3 and ILT4. Nat Immunol 3: 237–243.
  21. 21. Zhang L, Bertucci AM, Ramsey-Goldman R, Burt RK, Datta SK (2009) Regulatory T cell (Treg) subsets return in patients with refractory lupus following stem cell transplantation, and TGF-beta-producing CD8+ Treg cells are associated with immunological remission of lupus. J Immunol 183: 6346–6358.
  22. 22. Luo X, Tarbell KV, Yang H, Pothoven K, Bailey SL, et al. (2007) Dendritic cells with TGF-beta1 differentiate naive CD4+CD25− T cells into islet-protective Foxp3+ regulatory T cells. Proc Natl Acad Sci U S A 104: 2821–2826.
  23. 23. Saito M, Nagasawa M, Takada H, Hara T, Tsuchiya S, et al. (2011) Defective IL-10 signaling in hyper-IgE syndrome results in impaired generation of tolerogenic dendritic cells and induced regulatory T cells. J Exp Med 208: 235–249.
  24. 24. Bedke T, Pretsch L, Karakhanova S, Enk AH, Mahnke K (2010) Endothelial cells augment the suppressive function of CD4+ CD25+ Foxp3+ regulatory T cells: involvement of programmed death-1 and IL-10. J Immunol 184: 5562–5570.
  25. 25. Hippen KL, Merkel SC, Schirm DK, Nelson C, Tennis NC, et al. (2011) Generation and Large-Scale Expansion of Human Inducible Regulatory T Cells That Suppress Graft-Versus-Host Disease. Am J Transplant.
  26. 26. Chiu WK, Fann M, Weng NP (2006) Generation and growth of CD28nullCD8+ memory T cells mediated by IL-15 and its induced cytokines. J Immunol 177: 7802–7810.
  27. 27. Borthwick NJ, Lowdell M, Salmon M, Akbar AN (2000) Loss of CD28 expression on CD8(+) T cells is induced by IL-2 receptor gamma chain signalling cytokines and type I IFN, and increases susceptibility to activation-induced apoptosis. Int Immunol 12: 1005–1013.
  28. 28. Kapp JA, Honjo K, Kapp LM, Xu X, Cozier A, et al. (2006) TCR transgenic CD8+ T cells activated in the presence of TGFbeta express FoxP3 and mediate linked suppression of primary immune responses and cardiac allograft rejection. Int Immunol 18: 1549–1562.
  29. 29. He H, Yang P, Jiang L, Zhang J, Zhao C, et al. (2008) Upregulation of CD94 on CD8+T cells in anterior chamber-associated immune deviation. BMC Immunol 9: 53.
  30. 30. Hahn BH, Singh RP, La Cava A, Ebling FM (2005) Tolerogenic treatment of lupus mice with consensus peptide induces Foxp3-expressing, apoptosis-resistant, TGFbeta-secreting CD8+ T cell suppressors. J Immunol 175: 7728–7737.
  31. 31. Myers L, Croft M, Kwon BS, Mittler RS, Vella AT (2005) Peptide-specific CD8 T regulatory cells use IFN-gamma to elaborate TGF-beta-based suppression. J Immunol 174: 7625–7632.
  32. 32. Seo SK, Choi JH, Kim YH, Kang WJ, Park HY, et al. (2004) 4-1BB-mediated immunotherapy of rheumatoid arthritis. Nat Med 10: 1088–1094.
  33. 33. Martin AJ, McCarthy D, Waltenbaugh C, Goings G, Luo X, et al. (2010) Ethylenecarbodiimide-treated splenocytes carrying male CD4 epitopes confer histocompatibility Y chromosome antigen transplant protection by inhibiting CD154 upregulation. J Immunol 185: 3326–3336.
  34. 34. Koide J, Engleman EG (1990) Differences in surface phenotype and mechanism of action between alloantigen-specific CD8+ cytotoxic and suppressor T cell clones. J Immunol 144: 32–40.
  35. 35. Damle NK, Engleman EG (1989) Antigen-specific suppressor T lymphocytes in man. Clin Immunol Immunopathol 53: S17–24.
  36. 36. Surh CD, Sprent J (2008) Homeostasis of naive and memory T cells. Immunity 29: 848–862.
  37. 37. Zheng J, Liu Y, Qin G, Chan PL, Mao H, et al. (2009) Efficient induction and expansion of human alloantigen-specific CD8 regulatory T cells from naive precursors by CD40-activated B cells. J Immunol 183: 3742–3750.
  38. 38. Gilliet M, Liu YJ (2002) Generation of human CD8 T regulatory cells by CD40 ligand-activated plasmacytoid dendritic cells. J Exp Med 195: 695–704.
  39. 39. Cobbold SP, Adams E, Nolan KF, Regateiro FS, Waldmann H (2010) Connecting the mechanisms of T-cell regulation: dendritic cells as the missing link. Immunol Rev 236: 203–218.
  40. 40. Bisikirska B, Colgan J, Luban J, Bluestone JA, Herold KC (2005) TCR stimulation with modified anti-CD3 mAb expands CD8+ T cell population and induces CD8+CD25+ Tregs. J Clin Invest 115: 2904–2913.
  41. 41. Herold KC, Gitelman SE, Masharani U, Hagopian W, Bisikirska B, et al. (2005) A single course of anti-CD3 monoclonal antibody hOKT3gamma1(Ala-Ala) results in improvement in C-peptide responses and clinical parameters for at least 2 years after onset of type 1 diabetes. Diabetes 54: 1763–1769.
  42. 42. Allan SE, Crome SQ, Crellin NK, Passerini L, Steiner TS, et al. (2007) Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol 19: 345–354.
  43. 43. Morgan ME, van Bilsen JH, Bakker AM, Heemskerk B, Schilham MW, et al. (2005) Expression of FOXP3 mRNA is not confined to CD4+CD25+ T regulatory cells in humans. Hum Immunol 66: 13–20.
  44. 44. Sansom DM, Walker LS (2006) The role of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in regulatory T-cell biology. Immunol Rev 212: 131–148.
  45. 45. Fife BT, Griffin MD, Abbas AK, Locksley RM, Bluestone JA (2006) Inhibition of T cell activation and autoimmune diabetes using a B cell surface-linked CTLA-4 agonist. J Clin Invest 116: 2252–2261.
  46. 46. Qureshi OS, Zheng Y, Nakamura K, Attridge K, Manzotti C, et al. (2011) Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science 332: 600–603.
  47. 47. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, et al. (2008) CTLA-4 control over Foxp3+ regulatory T cell function. Science 322: 271–275.
  48. 48. Onishi Y, Fehervari Z, Yamaguchi T, Sakaguchi S (2008) Foxp3+ natural regulatory T cells preferentially form aggregates on dendritic cells in vitro and actively inhibit their maturation. Proc Natl Acad Sci U S A 105: 10113–10118.
  49. 49. Huang L, Baban B, Johnson BA 3rd, Mellor AL (2010) Dendritic cells, indoleamine 2,3 dioxygenase and acquired immune privilege. Int Rev Immunol 29: 133–155.