Donor CD8 cells play a pivotal role in preventing allogeneic marrow graft rejection, possibly by generating cytotoxic effectors needed to eliminate recipient T cells remaining after the pretransplant conditioning regimen or by producing cytokines needed to support the growth and differentiation of hematopoietic stem cells. In the present study, we assessed the role of donor T-cell cytotoxic effector function as a mechanism for eliminating recipient CD8 cells that cause marrow graft rejection in mice. The ability to prevent rejection was minimally affected by the presence of a defect in Fas ligand binding or by the absence of granzyme B but was severely affected by the absence of perforin. Doubly mutant perforin-deficient, Fas ligand-defective CD8 cells were completely unable to prevent rejection. Our results indicate first that recipient CD8 effectors responsible for causing marrow graft rejection are sensitive to cytotoxicity mediated by both perforin- and Fas-ligand-dependent mechanisms, and second that donor T cells must have at least one functional cytotoxic mechanism to prevent allogeneic marrow graft rejection.

© 1998 by The American Society of Hematology.

DONOR CD8 CELLS PLAY a pivotal role in preventing allogeneic marrow graft rejection in mice and are substantially more effective than donor CD4 cells.1 Based on the known function and specificity of CD8 cells and CD4 cells in primary allospecific T-cell populations,2 we suggested that the generation of T- cytotoxic responses against major histocompatibility complex (MHC) class I alloantigens or class I-restricted peptides was the most likely mechanism by which donor T cells inactivate or eliminate the recipient effectors responsible for causing rejection. This hypothesis was supported by results demonstrating that T cells from nontolerant donors were more effective for preventing rejection than T cells from F13 or chimeric donors4 that could not recognize recipient alloantigens.

We have now assessed the role of cytotoxic effector function as a mechanism by which donor T cells might prevent allogeneic marrow graft rejection. The cytotoxic effector function of T lymphocytes can be mediated by two well-characterized mechanisms.5-7 One of these involves cell surface interactions between Fas ligand expressed on activated cytotoxic lymphocytes (CTL) and Fas expressed on target cells. The other involves granule exocytosis, a process in which perforin facilitates the translocation of granzymes from CTL into target cells. Both mechanisms induce apoptosis in target cells. Additional less well-characterized mechanisms are involved in cytolytic effector function against certain types of target cells.8In the present study, we evaluated the effect of mutations in the Fas-ligand and perforin/granzyme pathways on the ability of donor CD8 cells to prevent allogeneic marrow graft rejection caused by recipient CD8 cells. In further experiments, we evaluated the ability of Fas ligand-defective perforin-deficient T cells to cause graft-versus-host disease (GVHD).

Mice.

C57BL/6J (B6; H2b, Ly5.2) males, B6.C-H2bm1 (bm1; Ly5.2) males and females, and BALB/cJ (H2d) females were purchased from the Jackson Laboratory (Bar Harbor, ME). Founders for the Ly5-congenic B6.SJL-Ly5aPtprcaPep3b(B6-Ly5.1) strain (provided by Dr David Myers, Sloan Kettering Institute, New York, NY) and for Fas ligand-defective B6Smn.C3H-Faslgld (gld), granzyme B-deficient B6, 129-Gzmbtm1Ley9 and perforin-deficient C57BL/6-Pfptm1Sdz10 strains (purchased from the Jackson Laboratory) were bred at the Fred Hutchinson Cancer Research Center (FHCRC; Seattle, WA). Perforin-deficient, gld-homozygous founder males10(provided by Prof Jurg Tschopp, Institute of Biochemistry, University of Lausanne, Epalinges, Switzerland) were backcrossed togld females, and the offspring were intercrossed to generate perforin-deficient gld progeny. In these mice, the neocassette that disrupts perforin expression is inserted at the exon 3 SphI site.11 Perforin-deficient, gld-homozygous mice were also generated by intercrossing the offspring from mating (gld × C57BL/6-Pfptm1Sdz) F1 males with C57BL/6-Pfptm1Sdz females. In these mice, the neo cassette that disrupts perforin expression is inserted at the exon 3 BstEII site.10 Mice were housed in groups of 5 under specific pathogen-free conditions with twice weekly cage changes and received sterilized chow and acidified water (pH 3.5) ad libitum. Four weeks after marrow transplantation, mice were transferred to conventional housing conditions. Experiments were reviewed and approved by the Institutional Animal Care and Use Committee of the FHCRC.

Screening for gld and perforin mutations.

For detection of gld and wild-type Fasl alleles, peripheral blood leukocyte (PBL) DNA was amplified respectively with 5′ primer TTTGAGGAATCTAAGACCC or TTTGAGGAATCTAAGACCT and 3′ primer ATAGCTGACCTGTTGGACC (94°C for 4 minutes, followed by 30 seconds at 94°C, 30 seconds at 58°C, and 45 seconds at 72°C for 35 cycles with 1.5 mmol/L MgCl2, 200 μmol/L dNTP, and 0.5 U Taq polymerase; AmpliTaq Gold; Perkin Elmer, Foster City, CA), each yielding a 200-bp product. For detection of neoinsertions at the perforin exon 3 Sph I site11 andBstEII site,10 PBL DNA was amplified, respectively, with 5′ primer ACCTCCACTCCACCTTGA and 3′ primer GATAAAGTGCGTGCCATAG or with 5′ primer AGTGTGAGTGCCAGGATTC and 3′ primer CGGGGATTGTTATTGTTC (94°C for 9 minutes, followed by 30 seconds at 94°C, 30 seconds at 60°C, and 90 seconds at 72°C for 35 cycles with 1.5 mmol/L MgCl2, 500 μmol/L dNTP, and 1.0 U Taq polymerase). The wild-type alleles, respectively, yield 115- and 199-bp products, whereas the mutated alleles yield 1,100- to 1,200-bp products.

Marrow transplantation.

Recipients 8 to 12 weeks of age were prepared by total body irradiation (TBI) in a single fraction from dual-opposed60Co sources at an exposure rate of 20 cGy/min on the day before transplantation. Marrow obtained by femur flush was depleted of T lymphocytes by rabbit complement (1:10)-mediated lysis using a mixture of antibodies specific for Thy-1.2, CD4, and CD8, as previously described.1 Nylon wool-nonadherent lymphocytes obtained from pooled mesenteric, retroperitoneal, femoral, axillary, and cervical lymph node suspensions were depleted of B cells by centrifugal immunoadherence12 on plastic petri dishes precoated with goat antibody against mouse Ig (10 μg/mL in pH 9.4 buffer, 18 hours at 4°C) or with CD45R/B220-specific antibody RA3-6B2 (rat IgG2a; PharMingen, San Diego, CA) or RA3-3A1/6.1 (rat IgM; hybridoma obtained from American Type Culture Collection [ATCC], Rockville, MD; 12.5 μg/mL bound to dishes coated with goat antibody against rat Ig). CD8-enriched T cells were isolated either by centrifugal immunoadherent depletion of CD4 cells on dishes precoated with antibody GK1.5 (rat IgG2b; ATCC; antibody from hybridoma culture supernatant bound to dishes coated with goat antibody against rat Ig) or by immunomagnetic positive selection with colloidal superparamagnetic microbeads conjugated with monoclonal rat antibody against murine CD8 (Miltenyi Biotec Inc, Sunnyvale, CA) used according to the manufacturer’s instructions. CD8-enriched populations isolated by negative selection contained 82% to 94% (mean, 88.5%) CD8 cells and 1.0% to 2.4% (mean, 1.5%) CD4 cells. CD8-enriched populations isolated by positive selection contained 83% to 96% (mean, 90.7%) CD8 cells and 3.8% to 10.6% (mean, 7.1%) CD4 cells. For experiments in which engraftment was an endpoint, graded numbers of CD8-enriched LN cells were added to grafts containing 5.0 × 106 T-cell-depleted marrow cells. For experiments in which GVHD was an endpoint, aliquots containing 1.0 × 107 CD3 cells from pooled nylon wool- nonadherent spleen and lymph node suspensions were added to grafts containing 40 × 106 T-cell-depleted marrow cells. Cell mixtures were injected into recipients via the lateral tail vein.

Assessment of chimerism and enumeration of peripheral blood B cells.

PBL were stained with fluorescein isothiocyanate (FITC)-conjugated CD3-specific antibody and with biotinylated antibody specific for Ly5.1 or H2Kb and analyzed as previously described.1 For each experiment, thresholds for delineating positive and negative cells were determined by staining samples from appropriate positive and negative controls. In the T-cell and myeloid gates, respectively, negative control samples showed 0.15% to 1.56% (mean, 0.65%) and 0.56% to 14.0% (mean, 2.5%) background staining, whereas positive control samples showed 97.6% to 100% (mean, 99.3%) and 93.1% to 100% (mean, 98.0%) stained cells. For enumeration of B cells, PBL were stained with FITC-conjugated CD3-specific antibody and biotinylated antibody specific for CD45RA (14.8, rat IgG2b; hybridoma obtained from ATCC) or with phycoerythrin (PE)-conjugated CD3-specific antibody (PharMingen) and FITC-conjugated CD45R/B220-specific antibody (PharMingen). B cells were enumerated as the percentage of CD45RA+ or CD45R+, CD3 cells in the lymphoid gate defined by forward and side scatter characteristics.

Ability of donor CD8 cells to prevent marrow graft rejection.

Preliminary experiments demonstrated that TBI exposures ≥650 cGy were sufficient to prevent rejection in bm1 (Ly5.2) recipients transplanted with 5.0 × 106 T-cell-depleted marrow cells from MHC class I-disparate B6-Ly5.1 donors, but rejection occurred in recipients prepared with 550 cGy TBI (data not shown). The ability to overcome rejection by increased pretransplant TBI in this model is characteristic of a process mediated by T cells1,4 and not by natural killer (NK) cells13,14 of the recipient. Rejection of the B6-Ly5.1 marrow was prevented in some recipients prepared with 550 cGy TBI when as few as 5.0 × 104 CD8-enriched B6 (Ly5.2) LN cells were added to the graft, and rejection was prevented in all recipients when 8.0 × 105 CD8 cells were added to the graft (Table 1). Under these conditions, engraftment of the donor marrow depends on the presence of T cells in the graft. In most engrafted recipients, T cells in the blood on day 28 after transplantation were derived predominantly from Ly5.1 marrow progenitors and not from mature Ly5.2 LN cells added to the graft, and the proportion of donor marrow-derived T cells increased progressively during the first 3 months after transplantation. Thus, the B6 CD8 cells added to the graft did not prevent maturation of B6-Ly5.1 marrow progenitors in the bm1 recipient thymus.

Table 1.

Ability of Donor CD8 Cells to Prevent Rejection of Marrow in Recipients With MHC Class I Disparity

Day CD8 Cells AddedPercentage of Donor Marrow-Derived Cells
T Lymphocytes Granulocytes
28  None  1, 0, 1, 0, 0  3, 2, 2, 4, 1  
 5.0 × 104 39, 89, 0, 1, 1 96, 99, 2, 6, 3  
 2.0 × 105 88, 0, 94, 89, 94  100, 2, 99, 99, 99  
 8.0 × 105 89, 87, 89, 86, 80  100, 100, 98, 100, 100  
61  None  0, 0, 0, 3, 1  1, 0, 1, 3, 2  
 5.0 × 104 72, 99, 0, 0, 0  93, 94, 0, 1, 1  
 2.0 × 105 89, 3, 99, 89  71, 4, 96, 79  
 8.0 × 105 99, 94, 84, 92, 96  99, 86, 73, 86, 90  
98  None  0, 0, 3, 1, 1 1, 2, 7, 2, 3  
 5.0 × 104 92, 99, 1, 0, 2 78, 97, 3, 2, 4  
 2.0 × 105 99, 0, 99, 99 97, 1, 97, 99  
 8.0 × 105 97, 98, 100, 99, 99  99, 100, 99, 100, 98 
Day CD8 Cells AddedPercentage of Donor Marrow-Derived Cells
T Lymphocytes Granulocytes
28  None  1, 0, 1, 0, 0  3, 2, 2, 4, 1  
 5.0 × 104 39, 89, 0, 1, 1 96, 99, 2, 6, 3  
 2.0 × 105 88, 0, 94, 89, 94  100, 2, 99, 99, 99  
 8.0 × 105 89, 87, 89, 86, 80  100, 100, 98, 100, 100  
61  None  0, 0, 0, 3, 1  1, 0, 1, 3, 2  
 5.0 × 104 72, 99, 0, 0, 0  93, 94, 0, 1, 1  
 2.0 × 105 89, 3, 99, 89  71, 4, 96, 79  
 8.0 × 105 99, 94, 84, 92, 96  99, 86, 73, 86, 90  
98  None  0, 0, 3, 1, 1 1, 2, 7, 2, 3  
 5.0 × 104 92, 99, 1, 0, 2 78, 97, 3, 2, 4  
 2.0 × 105 99, 0, 99, 99 97, 1, 97, 99  
 8.0 × 105 97, 98, 100, 99, 99  99, 100, 99, 100, 98 

Groups of 5 irradiated (550 cGy) bm1 (Ly5.2) recipients were transplanted with 5.0 × 106 T-cell–depleted B6-Ly5.1 marrow cells alone or with the indicated numbers of B6 (Ly5.2) CD8-enriched LN cells added to the graft. The percentage of donor marrow-derived T cells and granulocytes in the peripheral blood of recipients surviving on days 28, 61, and 97 after transplantation was determined by two-color staining with CD3- and Ly5.1-specific antibodies. Data indicate results for individual recipients.

Effect of cytotoxicity mutations on the ability of donor T cells to prevent marrow graft rejection.

In a series of seven experiments, we evaluated the effect of mutations in the Fas-ligand and perforin/granzyme pathways on the ability of donor CD8 cells to prevent rejection of B6-Ly5.1 marrow in irradiated bm1 recipients. Each experiment included negative controls with no donor T cells added to the marrow and positive controls with wild-type B6 CD8-enriched LN T cells added to the graft. In this model, approximately 1.0 × 105 wild-type B6 CD8 cells prevent rejection in 50% of the recipients (Table 2). The defect in Fas-ligand-mediated cytotoxicity caused by the gld mutation had only a minimal effect on the ability of donor T cells to prevent rejection. Although rejection occurred in all 15 recipients transplanted with grafts containing 5.0 × 104 CD8 cells enriched by negative selection (P ≅ .01 compared with wild-type B6 CD8 cells), rejection was prevented in all recipients transplanted with grafts containing 2.0 × 105 or 8.0 × 105gld CD8 cells. The absence of granzyme B in donor CD8 cells also had a minimal effect on their ability to prevent rejection. Although rejection occurred in all 14 recipients transplanted with grafts containing 5.0 × 104 CD8 cells enriched by negative selection (P ≅ .02 compared with wild-type B6 CD8 cells), rejection was prevented in 9 of 10 recipients transplanted with grafts containing 2.0 × 105 or 8.0 × 105 granzyme B-deficient CD8 cells.

Table 2.

Effect of Cytotoxicity Mutations on the Ability of Donor T Cells to Prevent Marrow Graft Rejection

T-Cell Phenotype No. Rejected/No. Evaluated (%)
5 × 104 CD82 × 105 CD8 8 × 105 CD82-3 × 106 CD8 1-2 × 107 CD8
Wild-type* 22/34 (65)  6/23 (26)  0/15 (0) 
Fas-ligand–defective (gld)  15/15 (100)  0/5 (0) 0/5 (0)  
Granzyme B-deficient  14/14 (100)  1/5 (20) 0/5 (0)  
Perforin-deficient 4/4 (100)  8/9 (89) 6/8 (75)  4/8 (50)  0/4 (0) 
Perforin-deficient/gld  5/5 (100)  5/5 (100) 5/5 (100)  11/11 (100)  
Perforin-deficient/gld     5/5 (100) 
T-Cell Phenotype No. Rejected/No. Evaluated (%)
5 × 104 CD82 × 105 CD8 8 × 105 CD82-3 × 106 CD8 1-2 × 107 CD8
Wild-type* 22/34 (65)  6/23 (26)  0/15 (0) 
Fas-ligand–defective (gld)  15/15 (100)  0/5 (0) 0/5 (0)  
Granzyme B-deficient  14/14 (100)  1/5 (20) 0/5 (0)  
Perforin-deficient 4/4 (100)  8/9 (89) 6/8 (75)  4/8 (50)  0/4 (0) 
Perforin-deficient/gld  5/5 (100)  5/5 (100) 5/5 (100)  11/11 (100)  
Perforin-deficient/gld     5/5 (100) 

Irradiated (550 cGy) bm1 recipients were transplanted with grafts containing 5.0 × 106 B6-Ly5.1 T-cell–depleted marrow cells and the indicated numbers of CD8-enriched LN cells from wild-type B6 donors or from donors with mutations in T-cell cytotoxicity. Data indicate the proportions (percentages) of survivors with less than 20% Ly5.1-positive granulocytes in the blood when last tested at 1 to 3 months after transplantation. All but 7 engrafted recipients were observed for at least 2 months after transplantation and had 35% to 100% (mean, 93.5%) donor marrow-derived granulocytes when last tested before being euthanized. Only 1 engrafted recipient had less than 50% donor marrow-derived granulocytes at the time of euthanasia. Recipients with rejection had 0% to 13% (mean, 2.52%) donor marrow-derived granulocytes when last tested before being euthanized. Rejection occurred in 39 of 40 (97%) recipients transplanted with marrow containing no added T cells. Data are summarized from a series of seven experiments.

*

In four experiments in which control CD8 cells were enriched by immunoadherent negative selection, rejection occurred in 14 of 24 (58%), 2 of 13 (15%), and in 0 of 5 (0%) recipients transplanted with grafts respectively containing 5.0 × 104, 2.0 × 105, or 8.0 × 105 T cells. By logistic regression, 6.8 × 104 LN CD8 cells were sufficient to prevent rejection in 50% of the recipients. In three experiments in which control CD8 cells were enriched by immunomagnetic positive selection, rejection occurred in 8 of 10 (80%), 4 of 10 (40%), and in 0 of 10 (0%) recipients transplanted with grafts respectively containing 5.0 × 104, 2.0 × 105, or 8.0 × 105 T cells. By logistic regression, 1.66 × 105 LN CD8 cells enriched by positive selection were needed to prevent rejection in 50% of the recipients (P = .04 as compared with LN CD8 cells enriched by negative selection). These results suggest that binding of immunomagnetic particles to CD8 molecules had a small effect on the ability of CD8 cells to prevent rejection, but this effect was easily overcome by increasing the number of CD8 cells added to the graft.

This strain had perforin expression disrupted by an insertion at the exon 3 BstEII site and was fully backcrossed to B6.

This strain had perforin expression disrupted by an insertion at the exon 3 Sph I site and was backcrossed to B6 for 8 to 10 generations.

Perforin-deficient donor CD8 cells were clearly impaired in their ability to prevent rejection. Results indicated that 2 to 3 × 106 perforin-deficient CD8 cells enriched by positive or negative selection were needed to prevent rejection in 50% of recipients. With 1.0 × 107 positively selected perforin-deficient CD8 cells added to the graft, rejection was prevented in all recipients. These results demonstrate that perforin-deficient CD8 cells have approximately 3% to 5% of wild-type activity in preventing marrow graft rejection in this model.

Prevention of marrow graft rejection by donor CD8 cells was tested with two strains having defects in both the Fas ligand and perforin pathways of cytotoxicity. Rejection was not prevented by as many as 2.0 × 107 positively selected CD8 cells from doubly mutant donors with perforin expression disrupted by an insertion at the exon 3Sph I site. Because this perforin-deficient strain was backcrossed to B6 for 8 to 10 generations, graft failure could have been caused through recognition of the donor B6-Ly5.1 marrow by the mutant T cells and not through failure to prevent rejection by the bm1 recipient. This issue was addressed in two ways. First, heavily irradiated (950 cGy) bm1 recipients were transplanted with grafts containing 5.0 × 106 T-cell-depleted B6-Ly5.1 marrow cells alone (n = 4) or with 1.0 × 107 CD8- enriched LN cells from (Sph I) perforin-deficientgld-homozygous donors added to the graft (n = 2). All recipients survived and were engrafted with Ly5.1-positive cells when tested at 28 and 61 days after transplantation (data not shown). These results demonstrate that rejection of the B6-Ly5.1 marrow in 550-cGy irradiated bm1 recipients could not have been caused by mutant T cells added to the graft. In a further experiment, we tested CD8 cells from doubly mutant donors generated from a perforin-deficient strain that was fully backcrossed to B6. Rejection was not prevented by 1.0 × 107 positively selected CD8 cells from doubly mutant donors with perforin expression disrupted by an insertion at the exon 3BstEII site (Table 2). Taken together, these results demonstrate that the doubly mutant T cells had less than 1% of wild-type activity in preventing marrow graft rejection in this model.

Ability of T cells with cytotoxicity mutations to cause GVHD.

Results of a previous study showed that T cells from donors with defects in both the Fas ligand and perforin pathways of cytotoxicity did not cause GVHD in sublethally irradiated recipients.8Our new findings demonstrating that donor T-cell-mediated cytotoxicity is necessary to prevent marrow graft rejection in sublethally irradiated recipients suggested that the absence of GVHD in the previous study could be explained by the lack of engraftment. Therefore, it was of interest to determine whether doubly mutant T cells could cause GVHD under conditions in which engraftment did not depend on the cytotoxic activity of these cells. For these experiments, lethally irradiated (1,100 cGy) BALB/c recipients were transplanted with 40 × 106 T-cell-depleted B6 or B6-Ly5.1 marrow cells alone or with 1.0 × 107 CD3 cells from doubly mutant donors added to the graft. All recipients were engrafted with donor cells, as demonstrated by H2 typing at 1 and 2 months after transplantation (data not shown). In each of three experiments, the perforin-deficient, Fas-ligand-defective T cells caused GVHD, as indicated by weight loss and B lymphopenia as compared with negative control recipients transplanted with marrow alone (Table 3). However, histopathologic examination of the skin, liver, gastrointestinal tract, and lungs with blinding of treatment groups showed no differences between mice transplanted with grafts containing mutant T cells and those transplanted with marrow alone. Recipients transplanted with grafts containing doubly mutant T cells did not have lymphadenopathy at the time of autopsy.

Table 3.

Manifestations of GVHD in Recipients Transplanted With Mutant T Cells

Experiment No. DayNo. Evaluated Body WeightPercentage of B Cells
Control MutantControl Mutant Control Mutant
1  91  5  101 ± 2.2  72 ± 8.5  53 ± 3.3  10 ± 6.0 
2  71  5  4  96 ± 2.0  74 ± 2.1 64 ± 2.5  23 ± 3.2  
3  70  4  105 ± 2.1  74 ± 6.4  59 ± 2.0 17 ± 5.0 
Experiment No. DayNo. Evaluated Body WeightPercentage of B Cells
Control MutantControl Mutant Control Mutant
1  91  5  101 ± 2.2  72 ± 8.5  53 ± 3.3  10 ± 6.0 
2  71  5  4  96 ± 2.0  74 ± 2.1 64 ± 2.5  23 ± 3.2  
3  70  4  105 ± 2.1  74 ± 6.4  59 ± 2.0 17 ± 5.0 

Groups of 5 lethally irradiated (1,100 cGy) BALB/c recipients were transplanted with grafts containing 40 × 106T-cell–depleted B6 or B6-Ly5.1 marrow cells alone (control) or with 1.0 × 107 CD3 cells from perforin-deficient, Fas-ligand–defective donors added to the graft (mutant). Body weight was measured as a percentage of the pretransplant baseline, and the percentage of B220+, CD3 cells in the blood lymphoid gate was measured by two-color flow cytometry among recipients surviving on the indicated day when the experiment was terminated. Shown are mean values ± 1 SEM for three consecutive experiments. In each experiment, the differences between groups were statistically significant. Experiments no. 1 and 3 were performed with mutant cells from mice, with perforin expression disrupted by an insertion at the exon 3 BstEII site. Experiment no. 2 was performed with mutant cells with perforin expression disrupted by an insertion at the exon 3Sph I site.

Results of the current study demonstrate that cytotoxicity mediated by either perforin- or Fas ligand-dependent mechanisms is necessary for enabling donor T cells to prevent allogeneic marrow graft rejection. Previous studies have shown that rejection of H-2K-disparate marrow is caused by recipient CD8 effectors that survive the pretransplant conditioning regimen.15 Our current results therefore indicate that these cells are sensitive to cytotoxicity mediated by both perforin or Fas ligand-dependent mechanisms. These results are consistent with observations that ConA blasts are sensitive to lysis by either mechanism but are not lysed by CTL in short-term (4 hours) assays when both mechanisms were absent.7,8 Even though activated CD8 cells are sensitive to apoptosis induced by ligation of TNFp75 receptors under certain conditions,16cytokine-mediated mechanisms are not sufficient for the elimination of recipient CD8 cells that cause marrow graft rejection.

Comparison of results with Fas-ligand defective CD8 cells and perforin-deficient CD8 cells suggests that the ability of donor T cells to prevent marrow graft rejection depends predominantly on perforin-dependent mechanisms of cytotoxicity. The striking difference in results between granzyme B-deficient and perforin- deficient T cells confirms that granzyme B is not essential for granule exocytosis mechanisms of cytotoxicity.17 In the absence of perforin, large numbers of donor CD8 cells were needed in order to prevent rejection, whereas Fas-ligand-defective CD8 cells were minimally impaired in their ability to prevent rejection. The relative importance of the perforin pathway as compared with the Fas-ligand pathway in cytotoxicity against T-cell targets has been demonstrated previously. Braun et al8 showed that sublethally irradiated perforin-deficient H2b recipients could not reject allogeneic H2d BALB/c spleen cell grafts and were therefore susceptible to lethal acute GVHD, whereas identically treated Fas-ligand-defective recipients did not develop GVHD, suggesting that the ability to prevent allogeneic T-cell engraftment was not impaired. These results are consistent with in vitro observations that the absence of Fas on ConA blast targets has limited effects on their susceptibility to lysis by CTL.7,8 

Baker et al18 have reported contrary results in experiments with B6 donors and lethally irradiated H2- compatible C3H.SW recipients. In their experiments, readily detectable numbers of host-derived myeloid cells, B lymphocytes, and thymocytes remained in recipients transplanted with grafts containing Fas-ligand-defective T cells, but few, if any, such host-derived cells were found in identically treated recipients transplanted with grafts containing perforin-deficient T cells. These results suggested that Fas-ligand-dependent mechanisms of cytotoxicity were predominantly responsible for eliminating recipient hematopoietic progenitors that survived the pretransplant conditioning regimen.

At least two explanations can be invoked to account for the differences in results among these studies. First, the targets killed by cytotoxic effectors were not identical in the two studies. T cells of the recipient or donor were the targets evaluated in our experiments, whereas hematopoietic progenitors of the recipient were the targets evaluated by Baker et al.18 Thus, T cells might be less susceptible to Fas-ligand-mediated cytotoxicity or more susceptible to perforin-mediated cytotoxicity as compared with hematopoietic progenitors. Second, the effectors evaluated in the two studies might not be identical. Although we have tested LN CD8 cells, the effectors responsible for elimination of recipient cells in the H2-identical B6 → C3H.SW transplant model have not been identified. If the minor histocompatibility antigens that cause GVHD in this model are presented by MHC-class II molecules, the effectors could be CD4 cells. This suggestion is consistent with observations that CD4 cells mediate cytotoxicity through Fas-ligand-dependent mechanisms and not through perforin-dependent mechanisms.19-21 

The availability of murine strains with induced mutations has facilitated considerable progress in dissecting the effector mechanisms contributing to GVHD.8,18,22 Previous studies have shown that perforin-deficient T cells have a decreased ability to induce GVHD as measured by the number of donor T cells required to cause mortality in recipients with disparity for major or minor histocompatibility antigens.8,22 Recipients transplanted with Fas-ligand-defective T cells from gld donors developed cachexia, but inflammatory lesions in the skin and liver were absent, and the profound B-lymphoid hypoplasia typically associated with GVHD did not occur.22 Our current results highlight the role of cytokine-mediated mechanisms by demonstrating that GVHD can occur in the absence of Fas-ligand- and perforin-mediated cytotoxicity. On the other hand, with a sufficient number of donor cells, lethal GVHD can also occur in the absence of signals transduced by ligation of TNFp55 receptors.23 Taken together, these results suggest that GVHD can develop both through cell-mediated cytotoxicity and through cytokine-mediated mechanisms. Our results confirm the earlier report by Baker et al,22 who showed that inflammatory cell infiltrates and apoptotic epithelial damage are not prominent in the liver and skin when GVHD is caused by cells that lack Fas-ligand.

Our current results demonstrating the importance of perforin-mediated cytotoxicity as a mechanism enabling donor T cells to prevent allogeneic marrow graft rejection suggest possible approaches for future clinical trials. Recent studies have shown that cytotoxic CD8 cells can have either a type 1 or type 2 cytokine profile.24,25 Type 1 polarized CD8 cells mediate cytotoxicity through either Fas-ligand or perforin-dependent mechanisms, whereas type 2 polarized CD8 cells rely predominantly on perforin-dependent mechanisms.26 Type 2 polarized CD8 cells also have a reduced ability to cause GVHD as compared with type 1 polarized cells.27,28 Taken together, these results suggest that retention of type 2 polarized donor CD8 cells in the graft could avoid marrow graft rejection when T-cell depletion is used to prevent GVHD.

The authors thank Kelli McIntyre and Natasha Leman for technical assistance and animal husbandry, Dr Wendy Leisenring for statistical consultation, Drs Ilonna Rimm and Claudio Anasetti for critical reading of the manuscript, and Alison Sell for assistance in preparing the manuscript.

Supported by US Public Health Service Grant No. HL-55257 awarded by the Department of Health and Human Services, National Institutes of Health.

Address correspondence to Paul J. Martin, MD, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D2-100, Seattle, WA 98109-1024; e-mail: pmartin@fhcrc.org.

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

1
Martin
PJ
Donor CD8 cells prevent allogeneic marrow graft rejection in mice: Potential implications for marrow transplantation in humans.
J Exp Med
178
1993
703
2
Golding
H
Mizuochi
T
McCarthy
SA
Cleveland
CA
Singer
A
Relationship among function, phenotype, and specificity in primary allospecific T cell populations: identification of phenotypically identical but functionally distinct primary T cell subsets that differ in their recognition of MHC class I and class II allodeterminants.
J Immunol
138
1987
10
3
Lapidot
T
Lubin
I
Terenzi
A
Faktorowich
Y
Erlich
P
Reisner
Y
Enhancement of bone marrow allografts from nude mice into mismatched recipients by T cells void of graft-versus-host activity.
Proc Natl Acad Sci USA
87
1990
4595
4
Martin
PJ
Prevention of allogenic marrow graft rejection by donor T cells that do not recognize recipient alloantigens: potential role of a veto mechanism.
Blood
88
1996
962
5
Kagi
D
Vignaux
F
Ledermann
B
Burki
K
Depraetere
V
Nagata
S
Hengartner
H
Golstein
P
Fas and perforin pathways as major mechanisms of T cell-mediated cytotoxicity.
Science
265
1994
528
6
Lowin
B
Hahne
M
Mattmann
C
Tschopp
J
Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways.
Nature
370
1994
650
7
Kojima
H
Shinohara
N
Hanaoka
S
Someya-Shirota
Y
Takagaki
Y
Ohno
H
Saito
T
Katayama
T
Yagita
H
Okumura
K
Shinkai
Y
Alt
FW
Matsuzawa
A
Yonehara
S
Takayama
H
Two distinct pathways of specific killing revealed by perforin mutant cytotoxic T lymphocytes.
Immunity
1
1994
357
8
Braun
MY
Lowin
B
French
L
Acha-Orbea
H
Tschopp
J
Cytotoxic T cells deficient in both functional Fas ligand and perforin show residual cytolytic activity yet lose their capacity to induce lethal acute graft- versus-host disease.
J Exp Med
183
1996
657
9
Heusel
JW
Wesselschmidt
RL
Shresta
S
Russell
JH
Ley
TJ
Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells.
Cell
76
1994
977
10
Kagi
D
Ledermann
B
Burki
K
Seiler
P
Odermatt
B
Olsen
KJ
Podack
ER
Zinkernagel
RM
Hengartner
J
Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforin-deficient mice.
Nature
369
1994
31
11
Lowin
B
Beermann
F
Schmidt
A
Tschopp
J
A null mutation in the perforin gene impairs cytolytic T lymphocyte- and natural killer cell-mediated cytotoxicity.
Proc Natl Acad Sci USA
91
1994
11571
12
Young
JW
Steinman
RM
Dendritic cells stimulate primary human cytolytic lymphocyte responses in the absence of CD4+ helper T cells.
J Exp Med
171
1990
1315
13
Cudkowicz
G
Bennett
M
Peculiar immunobiology of bone marrow allografts. I. Grafts rejection by irradiated responder mice.
J Exp Med
134
1971
83
14
Cudkowicz
G
Bennett
M
Peculiar immunobiology of bone marrow allografts. II. Rejection of parental grafts by resistant F1 hybrid mice.
J Exp Med
134
1971
1513
15
Vallera
DA
Taylor
PA
Sprent
J
Blazar
BR
The role of host T cell subsets in bone marrow rejection directed to isolated major histocompatibility complex class I versus class II differences of bm1 and bm12 mutant mice.
Transplantation
57
1994
249
16
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
17
Shresta
S
Russell
JH
Ley
TJ
Mechanisms responsible for granzyme B-independent cytotoxicity.
Blood
89
1997
4085
18
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
19
Hanabuchi
S
Koyanagi
M
Kawasaki
A
Shinohara
N
Matsuzawa
A
Nishimura
Y
Kobayashi
Y
Yonehara
S
Yagita
H
Okumura
K
Fas and its ligand in a general mechanism of T-cell-mediated cytotoxicity.
Proc Natl Acad Sci USA
91
1994
4930
20
Stalder
T
Hahn
S
Erb
P
Fas antigen is the major target molecule for CD4+ T cell-mediated cytotoxicity.
J Immunol
152
1994
1127
21
Ju
ST
Cui
H
Panka
DJ
Ettinger
R
Marshak-Rothstein
A
Participation of target Fas protein in apoptosis pathway induced by CD4+ Th1 and CD8+ cytotoxic T cells.
Proc Natl Acad Sci USA
91
1994
4185
22
Baker
MB
Altman
NH
Podack
ER
Levy
RB
The role of cell-mediated cytotoxicity in acute GVHD after MHC- matched allogeneic bone marrow transplantation in mice.
J Exp Med
183
1996
2645
23
Speiser
DE
Bachmann
MF
Frick
TW
McKall-Faienza
K
Griffiths
E
Pfeffer
K
Mak
TW
Ohashi
PS
TNF receptor p55 controls early acute graft-versus-host disease.
J Immunol
158
1997
5185
24
Croft
M
Carter
L
Swain
SL
Dutton
RW
Generation of polarized antigen-specific CD8 effector populations: Reciprocal action of interleukin (IL)-4 and IL-12 in promoting type 2 versus type 1 cytokine profiles.
J Exp Med
180
1994
1715
25
Sad
S
Marcotte
R
Mosmann
TR
Cytokine-induced differentiation of precursor mouse CD8+ T cells into cytotoxic CD8 T cells secreting Th1 or Th2 cytokines.
Immunity
2
1995
271
26
Carter
LL
Dutton
RW
Relative perforin- and Fas-mediated lysis in T1 and T2 CD8 effector populations.
J Immunol
155
1995
1028
27
Krenger
W
Snyder
KM
Byon
JCH
Falzarano
G
Ferrara
JLM
Polarized type 2 alloreactive CD4+ and CD8+ donor T cells fail to induce experimental acute graft-versus-host disease.
J Immunol
155
1995
585
28
Fowler
DH
Breglio
J
Nagel
G
Eckhaus
MA
Gress
RE
Allospecific CD8+ Tc1 and Tc2 populations in graft- versus-leukemia effect and graft-versus-host disease.
J Immunol
157
1996
4811
Sign in via your Institution