After hematopoietic stem cell transplantation, the persistence and expansion of grafted mature postthymic T cells allow both transfer of donor immunologic memory and generation of a diverse T repertoire. This thymic-independent process, which is particularly important in humans, because most transplant recipients present severe thymus atrophy, is impaired by graft-versus-host disease (GVHD). The goal of this study was to decipher how GVHD influences the fate of grafted postthymic T cells. Two major findings emerged. First, we found that, after a brisk proliferation phase, alloreactive antihost T cells underwent a massive activation-induced cell death (AICD). For both CD4+ and CD8+ T cells, the Fas pathway was found to play a major role in this AICD: alloreactive T cells upregulated Fas and FasL, and AICD of antihost T cells was much decreased in the case of lpr (Fas-deficient) donors. Second, whereas non–host-reactive donor T cells neither upregulated Fas nor suffered apoptosis when transplanted alone, they showed increased membrane Fas expression and apoptosis when coinjected with host-reactive T cells. We conclude that GVHD-associated AICD of antihost T cells coupled with bystander lysis of grafted non–host-reactive T cells abrogate immune reconstitution by donor-derived postthymic T lymphocytes. Furthermore, we speculate that massive lymphoid apoptosis observed in the acute phase of GVHD might be responsible for the occurrence of autoimmunity in the chronic phase of GVHD.

IN NORMAL INDIVIDUALS, the size of peripheral T-cell compartments is maintained by thymic and extrathymic pathways. First, thymic output is responsible for initial seeding of secondary lymphoid organs in young subjects.1 Continuous thymic output in later life contributes to the diversity of the T-cell repertoire,2,3 although the level of thymic function is known to decrease with age as the thymus undergoes progressive involution.4 Second, most T lymphocytes produced daily in adults derive from the proliferation of postthymic T cells that retain an impressive proliferative potential.5-7 Finally, extrathymic differentiation of hematopoietic progenitors has been detected in selected organs.8-12 The possible contribution of the latter pathway to T-cell homeostasis remains to be better defined but is generally believed to play a minor role compared with the first two other pathways.

It has been shown that, after transplantation of syngeneic hematopoietic stem cells, both thymic and extrathymic pathways can contribute to the reconstitution of peripheral T-cell compartments. In this model, the relative contribution of each pathway depends on the recipient’s thymic function and the number of postthymic T cells in the donor inoculum.13,14 In contrast, when the transplant is performed into an allogeneic recipient, graft-versus-host disease (GVHD) initiated by the graft postthymic T cells causes severe lymphoid hypoplasia.15-17 Disappearance of host lymphoid cells at the time of GVHD is easily accounted for by the generation of antihost alloreactive T cells that eradicate recipient mature lymphocytes and hematopoietic progenitors via both perforin and Fas-mediated cytotoxicity.16,18,19 However, there is no satisfactory explanation for the failure of donor-derived T cells to reconstitute GVHD+ hosts. It has been suggested that, during the course of GVHD, Fas/FasL interactions may contribute to failure of donor T-cell reconstitution, because donor T-cell hypoplasia is less severe when the donor is FasL defective or when the recipient is treated with anti-FasL monoclonal antibody (MoAb).16,20,21 However, interpretation of these interesting observations is limited by the fact that Fas/FasL interactions costimulate T-cell activation and proliferation in such a way that signaling through Fas in the early stages of an immune response augments the generation of effector cells.22,23 Indeed, after transplantation of FasL-defective cells, the generation of antihost effector cells has been shown to be severely impaired.20 Thus, it is not clear whether interference with Fas signaling decreases the severity of GVHD-related T-cell hypoplasia because (1) hypoplasia involves Fas-dependent apoptotic events or (2) because, similar to other molecules (eg, CD28, CD40, etc),24,25 the costimulatory effect of Fas is required for the expansion of alloreactive T cells.

The severity of GVHD-associated T-cell hypoplasia implies that GVHD abrogates both thymic and extrathymic T-cell reconstitution. Mechanistically, each differentiation pathway should be approached separately. It has been shown that the thymus stroma is a direct target of GVHD, and damage to the host thymus provides a rational explanation for the failure of donor-derived progenitors to differentiate via the classic central pathway.15,17,26 The problem in this case is probably one of soil rather than seed. In contrast, the failure of the graft mature postthymic T cells to expand and repopulate the host peripheral compartments, as observed in athymic syngeneic recipients, remains unexplained. This question is particularly relevant in the case of human recipients. Indeed, because of age- and disease-related factors such as chemotherapy, radiotherapy, or infections, most patients have thymus hypoplasia.27-31 In addition, T-cell reconstitution has been shown to depend mostly on proliferation of grafted postthymic T cells, at least during the first year posttransplant.32-34 Consequently, the failure of grafted postthymic T cells to expand in GVHD+ recipients can have a dramatic impact on immune reconstitution and is probably responsible to a large extent for the high frequency and severity of infections in these patients. Moreover, impairment of donor T-cell expansion could represent a major obstacle for the implementation of adoptive immunotherapy strategies. The latter point is increasingly important, because innovative approaches aimed at transferring activated/memory T cells specific for pathogens such as cytomegalovirus (CMV) and Epstein-Barr virus (EBV) hold great promise for the treatment of infections in immunocompromised hosts.35-37Thence, the goal of the present work was to determine the fate of grafted postthymic T cells in GVHD+ recipients and to understand why these mature donor T cells fail to repopulate the host’s peripheral T-cell compartments.

Mice.

The following strains of mice were purchased from the Jackson Laboratory (Bar Harbor, ME): A/J, C57BL/6J (B6), B6.SJL-PtprcaPep3b/BoyJ (Ly5a) (B6.SJL), B6.PL-Thy1a/Cy (Thy1.1) (B6.PL), B6.MRL-Faslpr (lpr), and B6Smn.C3H-Faslgld (gld). Mice with a B6 background have the H2b haplotype, whereas A/J mice are H2a. All inbred mice are Ly5.2+, except for B6.SJL mice, which are Ly5.1+. F1 hybrids from B6.SJL and A/J mice (B6.SAF1), and from B6 and A/J mice (B6AF1) were bred at the Guy-Bernier Research Center (Montreal, Quebec, Canada). Animals were maintained in specific pathogen-free conditions according to the standards of the Canadian Committee for Animal Protection. Lpr donors were between 5 and 8 weeks old, whereas other mice used as cell donors or recipients were between 5 and 16 weeks of age.

Induction of GVHD.

Spleen and lymph nodes (cervical, axillary, and inguinal) from donor mice with a B6 background were pooled into single-cell suspensions in which the number of Thy1+ T cells was assessed by flow cytometry. GVHD was induced by intravenous injection of a spleen/lymph node cell suspension containing 60 × 106 T cells into unirradiated F1 hybrid recipients.

Production of non–host-reactive cells.

Bone marrow cells were obtained from the tibiae and femurs of B6.PL donor mice and T-cell–depleted with specific anti–Thy-1 antiserum (Cedarlane, Hornby, Ontario, Canada) as previously described.13,38 Then, 107 bone marrow cells were intravenously injected into irradiated (12 Gy total body irradiation from a 60Co source at a dose rate of 128 cGy/min) B6AF1recipients on day 0. On day 60, spleen and lymph nodes of these B6AF1 recipients were harvested and used as a source of non–host-reactive cells.

MoAbs.

The following MoAbs were obtained from Pharmingen (San Diego, CA): fluorescein isothiocyanate (FITC)-labeled anti-Vβ panel, FITC- and phycoerythrin (PE)-labeled anti-Thy1.1 (MRC OX-7) and anti-Thy1.2 (53-2.1 and 30-H12, respectively), FITC-labeled and biotin-conjugated anti-Ly5.1 (A20) and anti-Ly5.2 (104), PE-labeled antibodies against CD19 (1D3), CD44 (1M7) and Fas (Jo2), Cy-chrome–labeled anti-CD4 (RM4-5) and anti-CD8 (53-7.7), purified anti-FasL (MFL3), biotin-conjugated goat-antihamster IgG, PE-conjugated streptavidin, and isotypic controls.

Flow cytometric analysis.

Cells were analyzed on a FACScalibur (Becton Dickinson, San Jose, CA) using the CellQuest program (Becton Dickinson) or on a FACScan (Becton Dickinson) using the Lysis II program (Becton Dickinson). Lymphocytes were gated by forward and side scatter, and fluorescence data were collected for 10,000 cells. Studies of selected T-cell populations were performed on 5,000 to 10,000 gated cells that were CD4+ or CD8+ and expressed the Ly5/Thy1 phenotype of interest. Direct immunofluorescence staining was performed as previously described.38 A sensitive three-step indirect staining method was used to assess the low-level expression of cell surface FasL. First, cells were stained with purifed anti-FasL Ab. After two washes, cells were incubated with biotinylated second-step reagent and then, after two additional washes, PE-conjugated streptavidin was added together with other selected fluorochrome-conjugated antibodies for three-color staining.

Measurement of FITC-labeled Annexin-V binding.

Apoptosis was analyzed by quantifying phosphatidylserine residues exposed on the cell membrane. Spleen cells were first stained with membrane-specific antibodies. After two washes with phosphate-buffered saline, 3 μL of recombinant FITC-labeled Annexin-V (Pharmingen) was added to cells resuspended in 100 μL of binding buffer (10 mmol/L HEPES/NaOH, pH 7.4, 140 mmol/L NaCl, 5 mmol/L CaCl2). After 15 minutes of incubation in the dark at room temperature, 500 μL of binding buffer was added and the samples were analyzed on a FACScan flow cytometer. In control samples, propidium iodide (2 μg/mL) staining was performed to help set the limit used to discriminate Annexin-V–positive and –negative cells. In some experiments, Annexin-V staining was performed after stimulation with Concanavalin A (Con A). Briefly, cells diluted at 2 × 106/mL in RPMI 1640 supplemented with 10% fetal calf serum, 100 U/mL penicillin G, 100 μg/mL streptomycin, 2 mmol/L L-glutamine, 1 mmol/L sodium pyruvate, 5 × 10−5mol 2-mercaptoethanol (2-ME), and 2 μg/mL of Concanavalin were incubated at 37°C in a humidified atmosphere of 5% CO2. After 40 hours of culture, cells were washed twice before antibodies and Annexin-V staining.

Statistical analysis.

Results for group means were compared using the Student’st-test.

Experimental model.

GVHD was induced by injecting a mixture of C57BL/6 or B6.SJL (H-2b) lymph node + spleen cells containing 6 × 107 T lymphocytes into unirradiated B6AF1 or B6.SAF1 recipients (H-2b/a). The origin of cells in chimeras was determined according to their Ly5 phenotype: C57BL/6J, A/J, and B6AF1 mice are Ly5.1Ly5.2+, B6.SJL are Ly5.1+Ly5.2, and B6.SAF1 mice are Ly5.1+Ly5.2+. We elected to use unirradiated recipients because irradiation per se can induce a number of effects that could have confounded our analyses. Indeed, irradiation can (1) increase Fas expression,39 (2) induce extrathymic T-cell development,40 (3) impair the function of thymus stromal cells,41 and (4) trigger the release of inflammatory cytokines.42 By using unirradiated recipients, we ensured that changes found in our chimeras were caused only by GVHD.

Expansion and disappearance of postthymic T cells during the course of GVHD.

After injection of B6.SJL cells into B6AF1 hosts, a transient increase of host lymphocytes was followed by total and irreversible disappearance of host B cells by day 12 (Fig 1A) and of host T cells between days 16 and 50 (Fig 1B). Between days 4 and 12, donor-derived T cells showed a notable proliferation, involving mainly the CD8+ subset (Fig 1B and D). This was followed by a conspicuous lymphoid hypoplasia of longer duration for T cells than for B cells. On day 100, (donor-derived) B-cell numbers had regained normal levels, whereas the numbers of (donor-derived) T cells were decreased by 70% compared with age-matched controls. These observations are concordant with the description of T-cell reconstitution reported by Hakim et al43 in a similar model of GVHD.

Fig. 1.

Expansion and disappearance of postthymic T cells during the course of GVHD. A cell suspension containing 6 × 107B6.SJL T cells was injected into unirradiated B6AF1 recipients. Shown is a time course evaluation of the numbers of host- and donor-derived B cells (A) and T cells (B) found in the spleen of GVHD+recipients. CD44 phenotype of host (C) and donor (D) T cells found in the spleen of GVHD+ recipients. Three-color staining was performed using PE-labeled anti-CD19, anti-Thy1.2, or anti-CD44; Cy-chrome–labeled anti-CD4 or anti-CD8; and FITC-labeled anti-Ly5.1 or anti-Ly5.2. Results are presented as the mean ± SD in (A) and (B); for the sake of clarity, only the mean is shown in (C) and (D). There were three to four mice per group.

Fig. 1.

Expansion and disappearance of postthymic T cells during the course of GVHD. A cell suspension containing 6 × 107B6.SJL T cells was injected into unirradiated B6AF1 recipients. Shown is a time course evaluation of the numbers of host- and donor-derived B cells (A) and T cells (B) found in the spleen of GVHD+recipients. CD44 phenotype of host (C) and donor (D) T cells found in the spleen of GVHD+ recipients. Three-color staining was performed using PE-labeled anti-CD19, anti-Thy1.2, or anti-CD44; Cy-chrome–labeled anti-CD4 or anti-CD8; and FITC-labeled anti-Ly5.1 or anti-Ly5.2. Results are presented as the mean ± SD in (A) and (B); for the sake of clarity, only the mean is shown in (C) and (D). There were three to four mice per group.

Close modal

T-cell proliferation can be observed after cognate interactions with antigen-presenting cells or after exposure of bystander cells to high concentrations of cytokines (such as interferon [IFN], interleukin-2 [IL-2], IL-6, and tumor necrosis factor [TNF]) produced by antigen-specific T cells.44-46 Acquisition of an activated/memory phenotype, characterized by upregulation of CD44, is observed in antigen-specific T cells after cognate interactions, but not in proliferating bystander T cells.44,47,48Nevertheless, at least in the context of some viral infections, T cells that are antigen-experienced, and thus have already upregulated CD44, may be more susceptible to bystander activation than naı̈ve (CD44lo) T cells.47 Analysis of the CD44 phenotype of expanded host- and donor-derived postthymic T-cell populations showed divergent phenotypes. At its time of maximal expansion (day 12), the population of donor-derived T cells was composed mainly of CD44hi cells, with very few CD44lo elements (Fig 1). In contrast, at its peak (day 8), expansion of recipient T cells involved mainly CD44locells, with a lesser increase in the number of CD44intcells and a notable decrease in the amount of CD44hielements. These results suggest that the transient expansion of host T cells was a bystander effect induced by the GVHD-associated cytokine storm, whereas donor T-cell proliferation was induced by cognate interactions with host antigens. Considering that we infused 6 × 107 donor T cells and that approximately 3% of T cells respond to a given allo-H2 haplotype,49 the grafted inoculum used in the preceding experiments contained ≤2 × 106 host-reactive T cells and approximately 5.8 × 107 non–host-reactive T cells. Taking into account that only a portion of infused T cells home to the spleen, host-reactive T cells (≤2 × 106) would have to expand more than 30-fold to account for the approximately 6 × 107donor T cells that were found in the spleen of day-12 recipients (Fig1). By comparison, the bystander expansion of host T cells was much more modest, because the number of host T cells increased to a maximum of twofold on day 8 (Fig 1).

Assessment of Vβ usage by CD4+ and CD8+ cell populations on day 12 showed that expansion of donor T cells involved all Vβ families tested. Indeed, the Vβ profile of expanded donor CD4+ T cells was similar to that of normal B6.SJL controls, whereas the Vβ usage by the expanded CD8+ subset showed only minor differences when compared with controls (Fig 2). These observations show that the cell expansion was of polyclonal origin and was not elicited by a superantigen.50,51 

Fig. 2.

Donor-derived postthymic T cells express a diverse Vβ repertoire. A cell suspension containing 6 × 107 B6.SJL T cells was injected into unirradiated B6AF1 recipients. Three-color staining was performed using the following antibodies: FITC-labeled anti-Vβ, Cy-chrome–labeled anti-CD4 or anti-CD8, and biotin-coupled anti-Ly5.1 plus Streptavidin-PE. Chimeras were studied on day +12. Results are presented as the mean ± SD (3 mice per group). *P< .05 when compared with B6.SJL controls.

Fig. 2.

Donor-derived postthymic T cells express a diverse Vβ repertoire. A cell suspension containing 6 × 107 B6.SJL T cells was injected into unirradiated B6AF1 recipients. Three-color staining was performed using the following antibodies: FITC-labeled anti-Vβ, Cy-chrome–labeled anti-CD4 or anti-CD8, and biotin-coupled anti-Ly5.1 plus Streptavidin-PE. Chimeras were studied on day +12. Results are presented as the mean ± SD (3 mice per group). *P< .05 when compared with B6.SJL controls.

Close modal
Expansion of donor T cells essentially involves host-reactive cells and is thus antigen-specific.

To evaluate more directly the importance of antigen-specific versus bystander proliferation in donor T-cell expansion, we produced and studied the fate of a donor (B6.PL) T-lymphocyte population selectively depleted in host (H2a)-reactive cells. These (Ly5.2+Thy1.1+) non–host-reactive T cells were harvested from the spleen and lymph nodes of B6AF1 mice that had been irradiated (12 Gy) and injected 60 days before with 107T-cell–depleted B6.PL bone marrow cells. After differentiation of B6.PL progenitors in the B6AF1 thymus, the lymphoid organs of these chimeras are repopulated with B6.PL T cells from which anti-H2a reactive elements have been deleted. As expected,13 the proliferative activity of these T cells was normal when assessed after in vitro stimulation with mitogens (Con A and phytohemagglutinin [PHA]) or third-party major histocompatibility complex (MHC) antigens (H2d; data not shown).

A cell suspension containing 4 × 107non–host-reactive T cells (of B6.PL origin) and 4 × 107 B6.SJL T cells (containing host-reactive T cells) was injected into B6.AF1 hosts, and we compared the expansion of these two donor T-cell populations. The rationale was that, if donor T-cell proliferation was host antigen-specific, B6.PL-derived non–host-reactive T cells would not proliferate and the donor T-cell compartment would contain almost exclusively B6.SJL cells. On the contrary, if donor T-cell proliferation were mainly due to bystander activation, the ratio of B6.PL vs. B6.SJL cells would approach 1:1. When assessed on days 8 and 12 posttransplant, the ratio of B6.PL versus B6.SJL cells was 1:99 (Table 1). At the time of maximal T-cell expansion, on day 12, the mean absolute numbers of B6.PL-versus B6.SJL-derived T cells found in the spleen of GVHD+ recipients were 61 × 106 and 0.5 × 106, respectively (data not shown). This indicates that donor T-cell expansion involved host-reactive cells and that the importance of bystander proliferation was negligible. Thus, the intense donor T-cell proliferation was consecutive to cognate interactions with host antigens and not the result of paracrine stimulation of bystander (non–host-reactive) T cells.

Table 1.

Importance of Antigen-Specific Versus Bystander Proliferation in Donor T-Cell Expansion

Donor Cell Origin
Ly5.1+Thy1.2+ (host-reactive +)Ly5.2+Thy1.1+ (host-reactive −)
Day 8  
 CD4  28%  0.8%  
 CD8  71%  0.2% 
 Total  99%  1%  
 
Day 12  
 CD4  33% 0.6%  
 CD8  66%  0.4%  
 Total  99% 1% 
Donor Cell Origin
Ly5.1+Thy1.2+ (host-reactive +)Ly5.2+Thy1.1+ (host-reactive −)
Day 8  
 CD4  28%  0.8%  
 CD8  71%  0.2% 
 Total  99%  1%  
 
Day 12  
 CD4  33% 0.6%  
 CD8  66%  0.4%  
 Total  99% 1% 

B6AF1 recipients (Ly5.2+Thy1.2+) received a cell suspension containing 4 × 107 B6.SJL T cells (containing host-reactive T cells) (Ly5.1+Thy1.2+) and 4 × 107Ly5.2+Thy1.1+ non–host-reactive T cells. Results show the mean (N = 3) proportion of donor-derived T cells that were Ly5.2+Thy1.1+ (non–host-reactive) versus Ly5.1+Thy1.2+. Non–host-reactive T cells were harvested from day-60 lethally irradiated B6AF1 recipients transplanted with 107 T-cell–depleted bone marrow cells from B6.PL donors. Trace amounts of B6AF1-derived T cells that could have contaminated the inoculum of non–host-reactive cells were not enumerated by the Ly5.2+Thy1.1+ phenotype.

Host-reactive cells show massive apoptosis involving Fas/FasL interactions.

The observation that brisk expansion of host-reactive T cells is followed by severe and prolonged lymphoid hypoplasia suggests that activation of donor T cells leads to massive activation-induced cell death (AICD). We therefore used Annexin-V staining to measure the number of apoptotic B6.SJL donor T cells in the spleen of B6AF1 hosts. The percentage of apoptotic T cells in fresh spleen cell suspensions reached 22% for the CD4+ subset on day 16 and 32% for the CD8+ subset on day 12 (Fig 3). These numbers of apoptotic cells are significantly higher than those observed in control B6.SJL T cells (3% to 4.5%). Because the in vivo clearance of apoptotic cells is very rapid,52,53 our results suggest that extensive amounts of donor T cells are most likely eliminated by apoptosis during the course of acute GVHD.

Fig. 3.

Host-reactive cells show massive apoptosis involving Fas/FasL interactions. (A) Proportion of apoptotic (Annexin-V–positive) T cells in F1 recipients injected with B6.SJL vs. lpr cells. The mean numbers of apoptotic CD4+/CD8+ T cells was 4.5%/3% in B6.SJL controls and 5.8%/3.4% in lpr controls (n = 3; data not shown). Results are expressed as the mean ± SD, n = 3. *P < .05, **P < .01, ***P < .005 relative to B6.SJL donors. (B) Histograms showing results for a representative F1 recipient studied on day 16 after injection of B6.SJL or lpr cells (shaded diagram). The clear histogram represents a negative control (untreated B6.SJL or lpr mouse).

Fig. 3.

Host-reactive cells show massive apoptosis involving Fas/FasL interactions. (A) Proportion of apoptotic (Annexin-V–positive) T cells in F1 recipients injected with B6.SJL vs. lpr cells. The mean numbers of apoptotic CD4+/CD8+ T cells was 4.5%/3% in B6.SJL controls and 5.8%/3.4% in lpr controls (n = 3; data not shown). Results are expressed as the mean ± SD, n = 3. *P < .05, **P < .01, ***P < .005 relative to B6.SJL donors. (B) Histograms showing results for a representative F1 recipient studied on day 16 after injection of B6.SJL or lpr cells (shaded diagram). The clear histogram represents a negative control (untreated B6.SJL or lpr mouse).

Close modal

With this in mind, we asked whether Fas/FasL interactions played a significant role in the apoptosis of donor T cells. We inferred that, if apoptosis was Fas-dependent, it should not occur when the donor is Fas- or FasL-deficient. In a first set of experiments, we determined that only the study of Fas-deficient (lpr), but not of FasL-deficient (gld) donors would be appropriate to address this question. Indeed, after injection into B6.SAF1 hosts, T cells from gld donors showed no measurable expansion and were rapidly eliminated (Fig 4). This observation might be explained by the fact that Fas/FasL interactions have a costimulatory effect on T-cell activation and generation of effector cells.20,22,23 Moreover, the failure of T cells from gld donors to proliferate demonstrates that, at least in our model, the absence of GVHD with gld donors may not be accepted as an evidence that Fas-mediated apoptosis (as opposed to Fas-mediated costimulation) plays a role in GVHD. In contrast to gld donors, cells of lpr donors expanded vigorously in recipients (Fig 4). Furthermore, from day 12 to 16, the number of lpr-derived CD8+ T cells increased, in contrast to CD8+ cells obtained from normal donors, which decreased. More importantly, the number of apoptotic donor-derived CD4+ and CD8+ T cells was strikingly decreased in the case of lpr donors as compared with normal donors (Fig 3). These observations suggest that Fas signaling plays a prominent role in the AICD of host-reactive T cells. However, it should be noted that, in the case of lpr donors, CD4+ T cells did not expand to the same extent as CD8+ cells between days 12 and 16 (Fig 4). Although the reason for this observation is unclear, it suggests that there may be differences in the mechanisms controlling CD4+versus CD8+ cells in this setting.

Fig. 4.

Influence of Fas/FasL interactions on donor T-cell expansion. Expansion of donor T cells obtained from B6.SJL, gld (FasL-deficient), or lpr (Fas-deficient) mice after transplantation in B6AF1 or B6.SAF1 recipients. Three-color staining was performed with the following fluorochrome-conjugated antibodies: anti-Ly5.1 or anti-Ly5.2, anti-CD4 or anti-CD8, and anti-Thy1.1 or anti-Thy1.2. On day 16, the number of donor-derived CD8+ T cells was greater with lpr donors than with B6.SJL donors (***P < .005).

Fig. 4.

Influence of Fas/FasL interactions on donor T-cell expansion. Expansion of donor T cells obtained from B6.SJL, gld (FasL-deficient), or lpr (Fas-deficient) mice after transplantation in B6AF1 or B6.SAF1 recipients. Three-color staining was performed with the following fluorochrome-conjugated antibodies: anti-Ly5.1 or anti-Ly5.2, anti-CD4 or anti-CD8, and anti-Thy1.1 or anti-Thy1.2. On day 16, the number of donor-derived CD8+ T cells was greater with lpr donors than with B6.SJL donors (***P < .005).

Close modal

We then analyzed the kinetics of Fas and FasL expression on T-cell subsets between days 8 and 16 in B6AF1 recipients of a B6.SJL graft. A significant upregulation of Fas expression was found in CD4+ and CD8+ host and donor T cells (Fig 5). Fas upregulation was most noticeable on donor CD4+ T cells and was of limited duration, being observed on day 8 but not later. In addition, a significant increase in the proportion of FasL+ cells was found specifically among the donor CD8+ T-cell subset (Fig 6). Donor CD4+ T cells showed only a minimal increase in FasL expression that did not reach significance. Upregulation of FasL on donor CD8+ T cells was present at the three time points studied from days 8 to 16. These results suggest that Fas-dependent AICD of donor T cells is initiated specifically by FasL+ CD8+ host-reactive T cells.

Fig. 5.

Expression of Fas on donor and host T-cell subsets. A cell suspension containing 6 × 107 B6.SJL T cells was injected into unirradiated B6AF1 recipients. (A) Increased Fas expression on day 8. Three-color staining was performed with the following antibodies: FITC-labeled anti-Ly5.1 or anti-Ly5.2, Cy-chrome–labeled anti-CD4 or anti-CD8, and PE-labeled anti-Fas (shaded histogram) or its isotypic control (clear histogram). Results from one representative experiment of three are shown. (B) Time course evaluation of Fas expression. Mean ± SD, N = 3; nd, not done. *P < .05, **P < .01, ***P < .005 relative to control.

Fig. 5.

Expression of Fas on donor and host T-cell subsets. A cell suspension containing 6 × 107 B6.SJL T cells was injected into unirradiated B6AF1 recipients. (A) Increased Fas expression on day 8. Three-color staining was performed with the following antibodies: FITC-labeled anti-Ly5.1 or anti-Ly5.2, Cy-chrome–labeled anti-CD4 or anti-CD8, and PE-labeled anti-Fas (shaded histogram) or its isotypic control (clear histogram). Results from one representative experiment of three are shown. (B) Time course evaluation of Fas expression. Mean ± SD, N = 3; nd, not done. *P < .05, **P < .01, ***P < .005 relative to control.

Close modal
Fig. 6.

Upregulation of FasL expression on CD8+ T cells. A cell suspension containing 6 × 107 B6.SJL T cells was injected into unirradiated B6AF1 recipients. (A) Increased FasL expression on donor (Ly5.1+) CD8+ T cells on day 8. Cells were first labeled with hamster anti-FasL antibody, then with biotin-coupled goat antihamster antibody, and lastly with PE-conjugated streptavidin together with FITC-labeled anti-Ly5.1 and Cy-chrome–labeled anti-CD4 or anti-CD8 antibodies; only the last two steps were performed in staining control. FasL staining is represented by a shaded histogram and staining control as a clear histogram. The value shown in each box represents the specific staining of FasL+ cells. Results from one representative experiment of three are shown. (B) Time course evaluation of FasL expression. Mean ± SD, N = 3. **P < .01, ****P< .001 relative to control.

Fig. 6.

Upregulation of FasL expression on CD8+ T cells. A cell suspension containing 6 × 107 B6.SJL T cells was injected into unirradiated B6AF1 recipients. (A) Increased FasL expression on donor (Ly5.1+) CD8+ T cells on day 8. Cells were first labeled with hamster anti-FasL antibody, then with biotin-coupled goat antihamster antibody, and lastly with PE-conjugated streptavidin together with FITC-labeled anti-Ly5.1 and Cy-chrome–labeled anti-CD4 or anti-CD8 antibodies; only the last two steps were performed in staining control. FasL staining is represented by a shaded histogram and staining control as a clear histogram. The value shown in each box represents the specific staining of FasL+ cells. Results from one representative experiment of three are shown. (B) Time course evaluation of FasL expression. Mean ± SD, N = 3. **P < .01, ****P< .001 relative to control.

Close modal
The influence of host-reactive cells on the fate of non–host-reactive cells.

Although AICD entails the disappearance of most, if not all, host-reactive T cells, what is the fate of non–host-reactive donor T cells? To evaluate this question, we compared the fate of non–host-reactive T cells when injected either alone or with host-reactive T cells. After injection of 2 × 107(B6.PL→B6AF1) non–host-reactive T cells into B6AF1 hosts, 3.8 × 106 Ly5.2+Thy1.1+ T cells were found in day-12 recipients (Fig 7A). Strikingly, the recovery of non–host-reactive T cells was decreased by approximately fivefold when 4 × 107 B6.SJL T cells were coinjected (Fig 7A). As a negative control, we verified that coinjection of syngeneic (B6.SAF1) T cells did not decrease the recovery of non–host-reactive T cells. Thus, clearance of non–host-reactive T cells was accelerated in the presence of host-reactive T cells. We hypothesized that two mechanisms could be responsible for the rapid demise of non–host-reactive T cells in the presence of host-reactive T cells: (1) competition for space between host-reactive and nonreactive T cells and (2) apoptosis of bystander cells induced by host-reactive T cells. Three series of observations showed characteristic features of apoptosis in non–host-reactive T cells when coinjected with host-reactive T cells, but not when injected alone: (1) upregulation of Fas expression (Fig 7B), (2) downregulation of Thy1 (Fig 7C; decreased expression of several cell surface membrane antigens such as Thy1 is one of the characteristic changes found on apoptotic cells54), and (3) increased numbers of Annexin-V–positive T cells (Fig 8). Increased numbers of Annexin-V–positive elements among non–host-reactive T cells coinjected with host reactive T cells were observed when analyses were performed on fresh recipients’ splenocyte suspensions, but became more impressive when analyses were performed on mitogen-stimulated T cells (Fig 8). In a control group, we observed that coinjection of syngeneic (B6.SAF1) T cells did not increase the proportion of apoptotic elements among non–host-reactive cells (Fig8B). These results lead us to contend that the failure of grafted non–host-reactive postthymic T cells to reconstitute the immune system of GVHD+ hosts is a consequence of bystander killing mediated by host-reactive effector T cells.

Fig. 7.

Accelerated clearance of non–host-reactive donor (Ly5.2+Thy1.1+) T cells when coinjected with host-reactive (Ly5.1+Thy1.2+) T cells. (A) Numbers of CD4+ and CD8+non–host-reactive T cells found in the spleen of B6AF1 recipients on day 12. Host-reactive and non–host-reactive cells were prepared as described for Fig 3A. Non–host-reactive T cells (20 × 106) were injected alone or with either 40 × 106 host-reactive T cells or 40 × 106syngeneic (B6.SAF1) T cells. N = 3; *P < .05 when compared with results in mice transplanted with non–host-reactive T cells alone or non–host-reactive T cells + syngeneic T cells. No differences were found between mice transplanted with non–host-reactive T cells alone versus non–host-reactive T cells + syngeneic T cells. (B) Non–host-reactive grafted CD4+ T cells overexpress Fas when coinjected with host-reactive T cells (right panel), but not when injected alone (left panel). Fas expression was assessed on day 8 as described in Fig 5. Results from one representative experiment of three are shown. (C) Downregulation of Thy1 expression was observed on CD4+ and CD8+ non–host-reactive grafted T cells when coinjected with host-reactive T cells (right panel), but not when injected alone (left panel). Two- and three-color staining was performed with the following fluorochrome-conjugated antibodies: anti-Ly5.2 or anti-Thy1.1, anti-Fas, and anti-CD4 or anti-CD8. Results from one representative experiment of three are shown.

Fig. 7.

Accelerated clearance of non–host-reactive donor (Ly5.2+Thy1.1+) T cells when coinjected with host-reactive (Ly5.1+Thy1.2+) T cells. (A) Numbers of CD4+ and CD8+non–host-reactive T cells found in the spleen of B6AF1 recipients on day 12. Host-reactive and non–host-reactive cells were prepared as described for Fig 3A. Non–host-reactive T cells (20 × 106) were injected alone or with either 40 × 106 host-reactive T cells or 40 × 106syngeneic (B6.SAF1) T cells. N = 3; *P < .05 when compared with results in mice transplanted with non–host-reactive T cells alone or non–host-reactive T cells + syngeneic T cells. No differences were found between mice transplanted with non–host-reactive T cells alone versus non–host-reactive T cells + syngeneic T cells. (B) Non–host-reactive grafted CD4+ T cells overexpress Fas when coinjected with host-reactive T cells (right panel), but not when injected alone (left panel). Fas expression was assessed on day 8 as described in Fig 5. Results from one representative experiment of three are shown. (C) Downregulation of Thy1 expression was observed on CD4+ and CD8+ non–host-reactive grafted T cells when coinjected with host-reactive T cells (right panel), but not when injected alone (left panel). Two- and three-color staining was performed with the following fluorochrome-conjugated antibodies: anti-Ly5.2 or anti-Thy1.1, anti-Fas, and anti-CD4 or anti-CD8. Results from one representative experiment of three are shown.

Close modal
Fig. 8.

Apoptosis of non–host-reactive T cells when coinjected with host-reactive T cells. Non–host-reactive cells were injected alone or with either host-reactive T cells or syngeneic T cells as described in Fig 7. (A) The proportion of non–host-reactive cells stained by Annexin-V was evaluated in fresh uncultured spleen cell suspensions from day-8 B6AF1 hosts. Three-color staining was performed with the following antibodies: PE-labeled anti-Ly5.2, Cy-chrome–labeled anti-CD4 or anti-CD8, and FITC-labeled Annexin-V. Results from one representative experiment of three are shown. (B) Annexin-V staining was performed after culturing recipient spleen cells during 40 hours in the presence of Con A. Results are expressed as the mean ± SD (N = 3) in the lower panel, and results from one representative experiment are depicted in the upper panel. **P< .01, ***P < .005 when compared with the results obtained in mice transplanted with non–host-reactive T cells alone or non–host-reactive T cells + syngeneic T cells. No differences were found between mice transplanted with non–host-reactive T cells alone versus non–host-reactive T cells + syngeneic T cells.

Fig. 8.

Apoptosis of non–host-reactive T cells when coinjected with host-reactive T cells. Non–host-reactive cells were injected alone or with either host-reactive T cells or syngeneic T cells as described in Fig 7. (A) The proportion of non–host-reactive cells stained by Annexin-V was evaluated in fresh uncultured spleen cell suspensions from day-8 B6AF1 hosts. Three-color staining was performed with the following antibodies: PE-labeled anti-Ly5.2, Cy-chrome–labeled anti-CD4 or anti-CD8, and FITC-labeled Annexin-V. Results from one representative experiment of three are shown. (B) Annexin-V staining was performed after culturing recipient spleen cells during 40 hours in the presence of Con A. Results are expressed as the mean ± SD (N = 3) in the lower panel, and results from one representative experiment are depicted in the upper panel. **P< .01, ***P < .005 when compared with the results obtained in mice transplanted with non–host-reactive T cells alone or non–host-reactive T cells + syngeneic T cells. No differences were found between mice transplanted with non–host-reactive T cells alone versus non–host-reactive T cells + syngeneic T cells.

Close modal
The fate of alloreactive antihost T cells.

After transplantation of T-cell–replete hematopoietic cell grafts, host-reactive T cells are initially sequestered in the spleen, where they proliferate before reentering the circulation and disseminating to other organs.55,56 By examining host spleen cell suspensions, we observed, in the early days posttransplant, a brisk expansion of alloreactive T cells followed by massive in situ AICD. Expanded populations of donor T cells comprised more CD8+than CD4+ T cells and were quite diverse in terms of Vβ repertoire. Expansion selectively involved antihost T cells. Non–host-reactive T cells did not expand when transplanted alone or with host-reactive cells. The latter observation indicates that T-cell proliferation triggered by host H2 antigens basically involves antigen-specific and not bystander T cells. Our finding is consistent with other recent studies on T-cell responses to viral antigens.46,57-61 

The very high numbers (up to 32%) of Annexin-V–labeled donor T cells found in fresh spleen cell suspensions from GVHD+ mice point to a massive GVHD-associated AICD. AICD is a physiologically important process that contributes to the termination of immune responses and can be mediated by numerous molecular pathways, including Fas, TNF, TRAIL, galectin, and CTLA-4.62-67 Analyses of T-cell responses to various antigens have shown that Fas usually plays a dominant role in AICD of CD4+ T cells.68-70The situation is more complex regarding AICD of CD8+ T cells. In the latter case, the role of Fas in various systems ranges from negligible to predominant.68,71-76 In models in which the Fas pathway seems to be of little significance, AICD is mediated predominantly by the TNF pathway.71,74 

Our results demonstrate the upregulation of FasL on CD8+host-reactive T cells, an increased expression of Fas on donor CD4+ and CD8+ T cells, and a major decrease in apoptotic donor CD4+ and CD8+ T cells in the case of lpr donors. They provide strong evidence that the Fas pathway plays a critical role in the AICD associated with GVHD. Furthermore, although it appears that both CD4+ and CD8+ T cells are targets of this AICD, effector function (FasL expression) seems to be performed only by host-reactive CD8+ T cells. Our data are concordant with results from others showing that the level of FasL upregulation detected on antigen reactive T cells after massive in vivo expansion is inferior to that observed on T cells after short-term in vitro culture with mitogens.18 This may well be explained by the recent observation that upregulation of FasL is an early and transient event that is maximal after one cell division but decreases after two to three cell divisions.77 Thus, when studies are performed on T-cell populations that have undergone massive in vivo expansion (here ≥30-fold for host-reactive T cells, ie, ≥5 cell divisions), FasL expression may not be at its maximum level. The molecular interactions responsible for the low levels of apoptosis observed in T cells from lpr donors remain to be defined. This could represent Fas-dependent apoptosis mediated by the low levels of Fas proteins expressed by lpr cells.78 Alternatively, residual levels of apoptosis detected with lpr donors could result from AICD mediated by TNF or other pathways and/or from passive cell death caused by deprivation of survival stimuli leading to decreased expression of antiapoptotic proteins, mainly of the Bcl family.64 

The fate of non–host-reactive T cells.

When injected alone, non–host-reactive T cells neither proliferated nor upregulated expression Fas expression. However, coinjection of host-reactive T cells entailed an increased expression of Fas on, in addition to apoptosis and accelerated disappearance of, non–host-reactive T cells. Therefore, apoptosis of non–host-reactive T cells and their subsequent failure to repopulate host secondary lymphoid organs likely represent collateral damage induced by FasL-expressing CD8+ host-reactive T cells. Bystander lysis of Fas+ T cells by FasL+ antigen-specific T cells has been reported in the course of viral infections.79-83 However, this collateral damage is usually of minimal significance because of its limited magnitude and because normal thymic output can replenish the peripheral T-cell pool. In contrast, we propose that, in the context of GVHD, bystander lysis of postthymic T cells can likely have far-reaching consequences. Indeed, severe thymic hypoplasia is commonly found in recipients of allogeneic hematopoietic stem cell transplants, particularly those with GVHD. Thymic-independent T-cell reconstitution via expansion of grafted postthymic T cells should be able to compensate for thymic failure.13,14 However, by abrogating this salvage pathway, bystander lysis of non–host-reactive donor T cells can dramatically compromise immune reconstitution.

Two different signaling pathways can increase Fas expression on T cells: TCR ligation and cytokines such as IFN-γ, IL-2, IL-7, and TNF-α.84-87 By definition, non–host-reactive T cells do not express antihost specific T-cell receptors. Thus, upregulation of Fas on these cells is presumably caused by secretion of IFN-γ, IL-2, IL-7, and TNF-α during the GVHD-associated cytokine storm.18,42,88,89 Interestingly, it has recently been shown in humans that acute GVHD, but not infection, is associated with increased soluble Fas serum levels.90 In the latter case, it remains to be determined whether increased serum Fas levels are caused by upregulation of Fas on donor T cells.

Patients with GVHD are profoundly immunodeficient. Therefore, they represent prime candidates for adoptive immunotherapy based on injection of donor-derived T cells specific for pathogens such as CMV or EBV.35-37 The results presented here suggest that, at least during the acute phase of GVHD, the brisk expansion of activated antihost CD8+ T cells can severely curtail the survival of transferred postthymic T cells. It will therefore be important to determine whether lysis of bystander postthymic T cells can be avoided by delaying adoptive transfer after the cytokine storm has subsided or after host-reactive T cells have gone through AICD.

Massive apoptosis: A link between GVHD and autoimmunity?

In normal individuals, apoptotic cells are readily cleared by the monocyte-macrophage system and do not elicit immune responses.52,53 However, under circumstances in which the clearance of apoptotic cells is impaired, it has been proposed that accumulation of high numbers of apoptotic cells could lead to immunogenic presentation of intracellular self-antigens and thereby initiate autoimmune responses.91,92 Direct evidence supporting this concept has recently been presented. Indeed, it has been shown that normal mice injected with large amounts of apoptotic syngeneic thymocytes (107 cells intravenously weekly for a total of 4 injections) develop a picture similar to that of systemic lupus erythematosus, characterized by autoantibodies and IgG deposition in the glomeruli.93 The investigators speculate that, when confronted with massive amounts of apoptotic (lymphoid) cells, the clearance capacity of macrophages could be overwhelmed and that abnormal (because of its level and/or duration) autoantigen presentation could pave the way to autoimmunity.93Additionally, studies in mice and humans have shown that chronic GVHD is more prevalent in individuals who have presented acute GVHD and shares many clinical, histologic, and immunologic features with autoimmune diseases such as systemic lupus erythematosus and Sjogren’s syndrome.94-96 How an alloimmune reaction (acute GVHD) can initiate autoimmunity (chronic GVHD) remains elusive. Our observations raise the possibility that massive apoptosis of donor T cells (via AICD and bystander lysis) could represent the missing link between these two processes.

Supported by the National Cancer Institute of Canada (C.P.). D.-C.R. is a senior scholar of the Fonds de la Recherche en Santé du Québec.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Modigliani
Y
Coutinho
G
Burlen-Defranoux
O
Coutinho
A
Bandeira
A
Differential contribution of thymic outputs and peripheral expansion in the development of peripheral T cell pools.
Eur J Immunol
24
1994
1223
2
Tanchot
C
Rocha
B
Peripheral selection of T cell repertoires: The role of continuous thymus output.
J Exp Med
186
1997
1099
3
Berzins
SP
Boyd
RL
Miller
JF
The role of the thymus and recent thymic migrants in the maintenance of the adult peripheral lymphocyte pool.
J Exp Med
187
1998
1839
4
George
AJ
Ritter
MA
Thymic involution with ageing: Obsolescence or good housekeeping?
Immunol Today
17
1996
267
5
Rocha
B
Dautigny
N
Pereira
P
Peripheral T lymphocytes: Expansion potential and homeostatic regulation of pool sizes and CD4/CD8 ratios in vivo.
Eur J Immunol
19
1989
905
6
Rocha
B
Penit
C
Baron
C
Vasseur
F
Dautigny
N
Freitas
AA
Accumulation of bromodeoxyuridine-labeled cells in central and peripheral lymphoid organs: Minimal estimates of production and turnover rates of mature lymphocytes.
Eur J Immunol
20
1990
1697
7
Sprent
J
Schaefer
M
Hurd
M
Surh
CD
Ron
Y
Mature murine B and T cells transferred to SCID mice can survive indefinitely and many maintain a virgin phenotype.
J Exp Med
174
1991
717
8
Dejbakhsh-Jones
S
Jerabek
L
Weissman
IL
Strober
S
Extrathymic maturation of αβ T cells from hemopoietic stem cells.
J Immunol
155
1995
3338
9
Garcia-Ojeda
ME
Dejbakhsh-Jones
S
Weissman
IL
Strober
S
An alternate pathway for T cell development supported by the bone marrow microenvironment: Recapitulation of thymic maturation.
J Exp Med
187
1998
1813
10
Saito
H
Kanamori
Y
Takemori
T
Nariuchi
H
Kubota
E
Takahashi-Iwanaga
H
Iwanaga
T
Ishikawa
H
Generation of intestinal T cells from progenitors residing in gut cryptopatches.
Science
280
1998
275
11
Sato
K
Ohtsuka
K
Hasegawa
K
Yamagiwa
S
Watanabe
H
Asakura
H
Abo
T
Evidence for extrathymic generation of intermediate T cell receptor cells in the liver revealed in thymectomized, irradiated mice subjected to bone marrow transplantation.
J Exp Med
182
1995
759
12
Clegg
CH
Rulffes
JT
Wallace
PM
Haugen
HS
Regulation of an extrathymic T-cell development pathway by oncostatin M.
Nature
384
1996
261
13
Dulude
G
Brochu
S
Fontaine
P
Baron
C
Gyger
M
Roy
DC
Perreault
C
Thymic and extrathymic differentiation and expansion of T lymphocytes following bone marrow transplantation in irradiated recipients.
Exp Hematol
25
1997
992
14
Mackall
CL
Gress
RE
Pathways of T-cell regeneration in mice and humans: Implications for bone marrow transplantation and immunotherapy.
Immunol Rev
157
1997
61
15
Lapp
WS
Ghayur
T
Mendes
M
Seddik
M
Seemayer
TA
The functional and histological basis for graft-versus-host-induced immunosuppression.
Immunol Rev
88
1985
107
16
Baker
MB
Riley
RL
Podack
ER
Levy
RB
Graft-versus-host-disease-associated lymphoid hypoplasia and B cell dysfunction is dependent upon donor T cell-mediated Fas-ligand function, but not perforin function.
Proc Natl Acad Sci USA
94
1997
1366
17
Hakim
FT
Mackall
CL
The immune system: Effector and target of graft-versus-host disease
Graft-vs.-Host Disease.
Ferrara
JLM
Deeg
HJ
Burakoff
SJ
1997
257
Marcel Dekker
New York, NY
18
Shustov
A
Nguyen
P
Finkelman
F
Elkon
KB
Via
CS
Differential expression of Fas and Fas ligand in acute and chronic graft-versus-host disease: up-regulation of Fas and Fas ligand requires CD8+ T cell activation and IFN-γ production.
J Immunol
161
1998
2848
19
Smit
WM
Rijnbeek
M
van Bergen
CAM
Fibbe
WE
Willemze
R
Falkenburg
JHF
T cells recognizing leukemic CD34+ progenitor cells mediate the antileukemic effect of donor lymphocyte infusions for relapsed chronic myeloid leukemia after allogeneic stem cell transplantation.
Proc Natl Acad Sci USA
95
1998
10152
20
Via
CS
Nguyen
P
Shustov
A
Drappa
J
Elkon
KB
A major role for the Fas pathway in acute graft-versus-host disease.
J Immunol
157
1996
5387
21
Hattori
K
Hirano
T
Miyajima
H
Yamakawa
N
Tateno
M
Oshimi
K
Kayagaki
N
Yagita
H
Okumura
K
Differential effects of anti-Fas ligand and anti-tumor necrosis factor α antibodies on acute graft-versus-host disease pathologies.
Blood
91
1998
4051
22
Lynch
DH
Ramsdell
F
Alderson
MR
Fas and FasL in the homeostatic regulation of immune responses.
Immunol Today
16
1995
569
23
Suzuki
I
Fink
PJ
Maximal proliferation of cytotoxic T lymphocytes requires reverse signaling through Fas ligand.
J Exp Med
187
1998
123
24
Blazar
BR
Korngold
R
Vallera
DA
Recent advances in graft-versus-host disease (GVHD) prevention.
Immunol Rev
157
1997
79
25
Yu
XZ
Martin
PJ
Anasetti
C
Role of CD28 in acute graft-versus-host disease.
Blood
92
1998
2963
26
Fukushi
N
Arase
H
Wang
B
Ogasawara
K
Gotohda
T
Good
RA
Onoe
K
Thymus: A direct target tissue in graft-versus-host reaction after allogeneic bone marrow transplantation that results in abrogation of induction of self-tolerance.
Proc Natl Acad Sci USA
90
1990
6301
27
Thomas
JA
Sloane
JP
Imrie
SF
Ritter
MA
Schuurman
HJ
Huber
J
Immunohistology of the thymus in bone marrow transplant recipients.
Am J Pathol
122
1986
531
28
Muller-Hermelink
HK
Sale
GE
Borisch
B
Storb
R
Pathology of the thymus after allogeneic bone marrow transplantation in man. A histologic immunohistochemical study of 36 patients.
Am J Pathol
129
1987
242
29
Mackall
CL
Fleisher
TA
Brown
MR
Andrich
MP
Chen
CC
Feuerstein
IM
Horowitz
ME
Magrath
IT
Shad
AT
Steinberg
SM
Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy.
N Engl J Med
332
1995
143
30
Mackall
CL
Fleisher
TA
Brown
MR
Andrich
MP
Chen
CC
Feuerstein
IM
Magrath
IT
Wexler
LH
Dimitrov
DS
Gress
RE
Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy.
Blood
89
1997
3700
31
Hakim
FT
Cepeda
R
Kaimei
S
Mackall
CL
McAtee
N
Zujewski
J
Cowan
K
Gress
RE
Constraints on CD4 recovery postchemotherapy in adults: Thymic insufficiency and apoptotic decline of expanded peripheral CD4 cells.
Blood
90
1997
3789
32
Roux
E
Helg
C
Dumont-Girard
F
Chapuis
B
Jeannet
M
Roosnek
E
Analysis of T-cell repopulation after allogeneic bone marrow transplantation: Significant differences between recipients of T-cell depleted and unmanipulated grafts.
Blood
87
1996
3984
33
Vavassori
M
Maccario
R
Moretta
A
Comoli
P
Wack
A
Locatelli
F
Lanzavecchia
A
Maserati
E
Dellabona
P
Casorati
G
Montagna
D
Restricted TCR repertoire and long-term persistence of donor-derived antigen-experienced CD4+ T cells in allogeneic bone marrow transplantation recipients.
J Immunol
157
1996
5739
34
Ottinger
HD
Beelen
DW
Scheulen
B
Schaefer
UW
Grosse-Wilde
H
Improved immune reconstitution after allotransplantation of peripheral blood stem cells instead of bone marrow.
Blood
88
1996
2775
35
Rooney
CM
Smith
CA
Ng
CY
Loftin
SK
Sixbey
JW
Gan
Y
Srivastava
DK
Bowman
LC
Krance
RA
Brenner
MK
Heslop
HE
Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients.
Blood
92
1998
1549
36
Walter
EA
Greenberg
PD
Gilbert
MJ
Finch
RJ
Watanabe
KS
Thomas
ED
Riddell
SR
Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor.
N Engl J Med
333
1995
1038
37
O’Reilly
RJ
Small
TN
Papadopoulos
E
Lucas
K
Lacerda
J
Koulova
L
Biology and adoptive cell therapy of Epstein-Barr virus-associated lymphoproliferative disorders in recipients of marrow allografts.
Immunol Rev
157
1997
195
38
Brochu
S
Baron
C
Hetu
F
Roy
DC
Perreault
C
Oligoclonal expansion of CTLs directed against a restricted number of dominant minor histocompatibility antigens in hemopoietic chimeras.
J Immunol
155
1995
5104
39
Reap
EA
Roof
K
Maynor
K
Borrero
M
Booker
J
Cohen
PL
Radiation and stress-induced apoptosis: A role for Fas/Fas ligand interactions.
Proc Natl Acad Sci USA
94
1997
5750
40
Lefrancois
L
Fuller
B
Huleatt
JW
Olson
S
Puddington
L
On the front lines: Intraepithelial lymphocytes as primary effectors of intestinal immunity.
Springer Semin Immunopathol
18
1997
463
41
Kumamoto
T
Inaba
M
Toki
J
Adachi
Y
Imamura
H
Ikehara
S
Cytotoxic effects of irradiation and deoxyguanosine on fetal thymus.
Immunobiology
192
1995
365
42
Antin
JH
Ferrara
JL
Cytokine dysregulation and acute graft-versus-host disease.
Blood
80
1992
2964
43
Hakim
FT
Sharrow
SO
Payne
S
Shearer
GM
Repopulation of host lymphohematopoietic systems by donor cells during graft-versus-host reaction in unirradiated adult F1 mice injected with parental lymphocytes.
J Immunol
146
1991
2108
44
Unutmaz
D
Pileri
P
Abrignani
S
Antigen-independent activation of naive and memory resting T cells by a cytokine combination.
J Exp Med
180
1994
1159
45
Tough
DF
Sprent
J
Viruses and T cell turnover: Evidence for bystander proliferation.
Immunol Rev
150
1996
129
46
Ehl
S
Hombach
J
Aichele
P
Hengartner
H
Zinkernagel
RM
Bystander activation of cytotoxic T cells: Studies on the mechanism and evaluation of in vivo significance in a transgenic mouse model.
J Exp Med
185
1997
1241
47
Tough
DF
Sprent
J
Turnover of naive- and memory-phenotype T cells.
J Exp Med
179
1994
1127
48
Sprent
J
Tough
DF
Lymphocyte life-span and memory.
Science
265
1994
1395
49
Ryser
JE
MacDonald
HR
Limiting dilution analysis of alloantigen-reactive T lymphocytes. I. Comparison of precursor frequencies for proliferative and cytolytic responses.
J Immunol
122
1979
1691
50
Held
W
Acha-Orbea
H
MacDonald
HR
Waanders
GA
Superantigens and retroviral infection: Insights from mouse mammary tumor virus.
Immunol Today
15
1994
184
51
Aoki
Y
Hiromatsu
K
Usami
J
Makino
M
Igarashi
H
Ogasawara
J
Nagata
S
Yoshikai
Y
Clonal expansion but lack of subsequent clonal deletion of bacterial superantigen-reactive T cells in murine retroviral infection.
J Immunol
153
1994
3611
52
Savill
J
Recognition and phagocytosis of cells undergoing apoptosis.
Br Med Bull
53
1997
491
53
Savill
J
Apoptosis. Phagocytic docking without shocking.
Nature
392
1998
442
54
Philippe
J
Louagie
H
Thierens
H
Vral
A
Cornelissen
M
De Ridder
L
Quantification of apoptosis in lymphocyte subsets and effect of apoptosis on apparent expression of membrane antigens.
Cytometry
29
1997
242
55
Korngold
R
Sprent
J
Selection of cytotoxic T-cell precursors specific for minor histocompatibility determinants. I. Negative selection across H-2 barriers induced with disrupted cells but not with glutaraldehyde-treated cells: Evidence for antigen processing.
J Exp Med
151
1980
314
56
Korngold
R
Sprent
J
Negative selection of T cells causing lethal graft-versus-host disease across minor histocompatibility barriers. Role of the H-2 complex.
J Exp Med
151
1980
1114
57
Zarozinski
CC
Welsh
RM
Minimal bystander activation of CD8 T cells during the virus-induced polyclonal T cell response.
J Exp Med
185
1997
1629
58
Butz
EA
Bevan
MJ
Massive expansion of antigen-specific CD8+ T cells during an acute virus infection.
Immunity
8
1998
167
59
Murali-Krishna
K
Altman
JD
Suresh
M
Sourdive
DJD
Zajac
AJ
Miller
JD
Slansky
J
Ahmed
R
Counting antigen-specific CD8 T cells: A reevaluation of bystander activation during viral infection.
Immunity
8
1998
177
60
Callan
MF
Tan
L
Annels
N
Ogg
GS
Wilson
JD
O’Callaghan
CA
Steven
N
McMichael
AJ
Rickinson
AB
Direct visualization of antigen-specific CD8+ T cells during the primary immune response to Epstein-Barr virus in vivo.
J Exp Med
187
1998
1395
61
Kuroda
MJ
Schmitz
JE
Barouch
DH
Craiu
A
Allen
TM
Sette
A
Watkins
DI
Forman
MA
Letvin
NL
Analysis of Gag-specific cytotoxic T lymphocytes in simian immunodeficiency virus-infected rhesus monkeys by cell staining with a tetrameric major histocompatibility complex class I-peptide complex.
J Exp Med
187
1998
1373
62
Perillo
NL
Pace
KE
Seilhamer
JJ
Baum
LG
Apoptosis of T cells mediated by galectin-1.
Nature
378
1995
736
63
Scheipers
P
Reiser
H
Fas-independent death of activated CD4+ T lymphocytes induced by CTLA-4 crosslinking.
Proc Natl Acad Sci USA
95
1998
10083
64
Van Parijs
L
Abbas
AK
Homeostasis and self-tolerance in the immune system: Turning lymphocytes off.
Science
280
1998
243
65
Van Parijs
L
Peterson
DA
Abbas
AK
The Fas/Fas ligand pathway and bcl-2 regulate T cell responses to model self and foreign antigens.
Immunity
8
1998
265
66
Van Parijs
L
Biuckians
A
Abbas
AK
Functional roles of fas and bcl-2-regulated apoptosis of T lymphocytes.
J Immunol
160
1998
2065
67
Martinez-Lorenzo
MJ
Alava
MA
Gamen
S
Kim
KJ
Chuntharapai
A
Pineiro
A
Naval
J
Anel
A
Involvement of APO2 ligand/TRAIL in activation-induced death of Jurkat and human peripheral blood T cells.
Eur J Immunol
28
1998
2714
68
Zheng
L
Fisher
G
Miller
RE
Peschon
J
Lynch
DH
Lenardo
MJ
Induction of apoptosis in mature T cells by tumour necrosis factor.
Nature
377
1995
348
69
Sytwu
HK
Liblau
RS
McDevitt
HO
The roles of Fas/APO-1 (CD95) and TNF in antigen-induced programmed cell death in T cell receptor transgenic mice.
Immunity
5
1996
17
70
Refaeli
Y
Van Parijs
L
London
CA
Tschopp
J
Abbas
AK
Biochemical mechanisms of IL-2-regulated Fas-mediated T cell apoptosis.
Immunity
8
1998
615
71
Ehl
S
Hoffmann-Rohrer
U
Nagata
S
Hengartner
H
Zinkernagel
R
Different susceptibility of cytotoxic T cells to CD95 (Fas/Apo-1) ligand-mediated cell death after activation in vitro versus in vivo.
J Immunol
156
1996
2357
72
Zimmermann
C
Rawiel
M
Blaser
C
Kaufmann
M
Pircher
H
Homeostatic regulation of CD8+ T cells after antigen challenge in the absence of Fas (CD95).
Eur J Immunol
26
1996
2903
73
Speiser
DE
Sebzda
E
Ohteki
T
Bachmann
MF
Pfeffer
K
Mak
TW
Ohashi
PS
Tumor necrosis factor receptor p55 mediates deletion of peripheral cytotoxic T lymphocytes in vivo.
Eur J Immunol
26
1996
3055
74
Lohman
BL
Razvi
ES
Welsh
RM
T-lymphocyte downregulation after acute viral infection is not dependent on CD95 (Fas) receptor-ligand interactions.
J Virol
70
1996
8199
75
Kurts
C
Heath
WR
Kosaka
H
Miller
JP
Carbone
FR
The peripheral deletion of autoreactive CD8+ T cells induced by cross-presentation of self-antigens involves signaling through CD95 (Fas, apo-1).
J Exp Med
188
1998
415
76
Mehal
WZ
Crispe
IN
TCR ligation on CD8+ T cells creates double-negative cells in vivo.
J Immunol
161
1998
1686
77
Oehen
S
Brduscha-Riem
K
Differentiation of naive CTL to effector and memory CTL: Correlation of effector function with phenotype and cell division.
J Immunol
161
1998
5338
78
Booker
JK
Reap
EA
Cohen
PL
Expression and function of fas on cells damaged by γ-irradiation in B6 and B6/lpr mice.
J Immunol
161
1998
4536
79
Kojima
H
Eshima
K
Takayama
H
Sitkovsky
MV
Leukocyte function-associated antigen-1-dependent lysis of Fas+ (CD95+/Apo-1+) innocent bystanders by antigen-specific CD8+ CTL.
J Immunol
159
1997
2728
80
Ando
K
Hiroishi
K
Kaneko
T
Moriyama
T
Muto
Y
Kayagaki
N
Yagita
H
Okumura
K
Imawari
M
Perforin, Fas/Fas ligand, and TNF-α pathways as specific and bystander killing mechanisms of hepatitis C virus-specific human CTL.
J Immunol
158
1997
5283
81
Hornung
F
Zheng
L
Lenardo
MJ
Maintenance of clonotype specificity in CD95/Apo-1/Fas-mediated apoptosis of mature T lymphocytes.
J Immunol
159
1997
3816
82
Smyth
MJ
Krasovskis
E
Johnstone
RW
Fas ligand-mediated lysis of self bystander targets by human papillomavirus-specific CD8+ cytotoxic T lymphocytes.
J Virol
72
1998
5948
83
Combadière
B
Reis e Sousa
C
Trageser
C
Zheng
LX
Kim
CH
Lenardo
MJ
Differential TCR signaling regulates apoptosis and immunopathology during antigen responses in vivo.
Immunity
9
1998
305
84
Itoh
N
Yonehara
S
Ishii
A
Yonehara
M
Mizushima
S
Sameshima
M
Hase
A
Seto
Y
Nagata
S
The polypeptide encoded by the cDNA for human cell surface antigen Fas can mediate apoptosis.
Cell
66
1991
233
85
Owen-Schaub
LB
Yonehara
S
Crump
WL
Grimm
EA
DNA fragmentation and cell death is selectively triggered in activated human lymphocytes by Fas antigen engagement.
Cell Immunol
140
1992
197
86
Moulian
N
Bidault
J
Planché
C
Berrih-Aknin
S
Two signaling pathways can increase Fas expression in human thymocytes.
Blood
92
1998
1297
87
Sayers
TJ
Brooks
AD
Lee
JK
Fenton
RG
Komschlies
KL
Wigginton
JM
Winkler-Pickett
R
Wiltrout
RH
Molecular mechanisms of immune-mediated lysis of murine renal cancer: Differential contributions of perforin-dependent versus Fas-mediated pathways in lysis by NK and T cells.
J Immunol
161
1998
3957
88
Levy
RB
Jones
M
Hamilton
BL
Paupe
J
Horowitz
T
Riley
R
IL-7 drives donor T cell proliferation and can costimulate cytokine secretion after MHC-matched allogeneic bone marrow transplantation.
J Immunol
154
1995
106
89
Krenger
W
Hill
GR
Ferrara
JLM
Cytokine cascades in acute graft-versus-host disease.
Transplantation
64
1997
553
90
Liem
LM
van Lopik
T
van Nieuwenhuijze
AE
van Houwelingen
HC
Aarden
L
Goulmy
E
Soluble fas levels in sera of bone marrow transplantation recipients are increased during acute graft-versus-host disease but not during infections.
Blood
91
1998
1464
91
Carroll
MC
The lupus paradox.
Nat Genet
19
1998
3
92
Rovere
P
Vallinoto
C
Bondanza
A
Crosti
MC
Rescigno
M
Ricciardi-Castagnoli
P
Rugarli
C
Manfredi
AA
Cutting edge: Bystander apoptosis triggers dendritic cell maturation and antigen-presenting function.
J Immunol
161
1998
4467
93
Mevorach
D
Zhou
JL
Song
X
Elkon
KB
Systemic exposure to irradiated apoptotic cells induces autoantibody production.
J Exp Med
188
1998
387
94
Parkman
R
Clonal analysis of murine graft-vs-host disease. I. Phenotypic and functional analysis of T lymphocyte clones.
J Immunol
136
1986
3543
95
Via
CS
Shearer
GM
T-cell interactions in autoimmunity: Insights from a murine model of graft-versus-host disease.
Immunol Today
9
1988
207
96
Siadak
M
Sullivan
KM
The management of chronic graft-versus-host disease.
Blood Rev
8
1994
154

Author notes

Address reprint requests to Claude Perreault, MD, Guy-Bernier Research Center, Maisonneuve-Rosemont Hospital, 5415 de l’Assomption Blvd, Montreal, Quebec, Canada H1T 2M4; e-mail: c.perreault@videotron.ca.

Sign in via your Institution