Abstract
In allogeneic hematopoietic stem cell transplantation, mature donor αβ T cells in the allograft promote T cell reconstitution in the recipient and mediate the graft-versus-leukemia (GVL) effect. Unfortunately, donor T cells can attack nonmalignant host tissues and cause graft-versus-host disease (GVHD). It has previously been shown that effector memory T cells not primed to alloantigen do not cause GVHD, yet transfer functional T cell memory and mediate GVL. Recently, central memory T cells (TCM) have also been reported to not cause GVHD. In contrast, here we demonstrate that purified CD8+ TCM not specifically primed to alloantigens mediate GVHD in the major histocompatibility complex (MHC)-mismatched B6→BALB/c and the MHC-matched, multiple minor histocompatibility antigen-mismatched C3H.SW→B6 strain pairings. CD8+ TCM and naïve T cells (TN) caused similar histologic disease in liver, skin and bowel. B6 CD8+ TCM and TN similarly expanded in BALB/c recipients and the majority of their progeny produced IFN-γ upon restimulation. However, in both models, CD8+ TCM induced milder clinical GVHD than did CD8+ TN. Nonetheless, CD8+ TCM and TN were similarly potent mediators of GVL against a mouse model of chronic phase chronic myelogenous leukemia. Thus, in contrast to what was previously thought, CD8+ TCM are capable of inducing GVHD, are substantially different from TEM, but only subtly so from TN.
Keywords: Graft Versus Host Disease, T cells, Tumor Immunity
Introduction
Allogeneic hematopoietic stem cell transplantation (alloSCT) is a potentially curative therapy for hematologic malignancies, including acute and chronic leukemias and lymphomas. In an alloSCT, donor T cells in the allograft are critical for reconstituting T cell immunity in the host (1) and mediate the graft-versus-leukemia (GVL) effect (2–5). Unfortunately, donor T cells also cause graft-versus-host disease (GVHD), the broad attack by donor T cells on recipient tissues, including the skin, liver and gastrointestinal tract. All clinical transplant protocols incorporate some type of GVHD prophylaxis, either via depletion of T cells from the allograft or through pharmacologic agents that impair T cell function. This GVHD prophylaxis impairs both GVL and immune reconstitution, and even with pharmacologic immunosuppression, GVHD remains a major source of morbidity and mortality. Preserving the positive effects of donor T cells—GVL and immune reconstitution—without GVHD remains the central challenge in the alloSCT field.
Peripheral T cells can broadly be divided into those that have never been activated by antigen (naïve T cells) and antigen experienced T cells, which include effector and memory T cells (6–9). Memory T cells are themselves heterogeneous and can be subdivided into effector memory (TEM) and central memory (TCM) fractions by surface phenotype and by function (10). TEM (CD62Llo/CCR7−CD44+) quickly express effector functions upon restimulation, preferentially migrate to inflamed tissues and the spleen, and are relatively excluded from peripheral lymph nodes (LN). TCM (CD62L+CCR7+CD44+) have hybrid properties of both TN and TEM. Like TN, their expression of CD62L and CCR7 promotes efficient homing to peripheral LNs (8, 10–16). However, while their effector functions are more vigorous than TN, they are slower to mount these responses than are TEM. Nonetheless, when rechallenged with antigen, TCM have a greater proliferative capacity than do TEM, and such a property could be advantageous in both anti-pathogen and anti-tumor responses (11, 17–21).
We and others have previously reported that CD4+ and CD8+ TEM have a reduced capacity to induce GVHD (22–26). CD4+ TEM are nonetheless functional and can transfer immunity to a model antigen and retain some GVL activity (22, 26). However, much less is known about the GVHD- and GVL-inducing potential of CD8+ TCM. Chen and colleagues reported that sorted CD45RB+CD62L+CD44+ TCM (a mix of CD4+ and CD8+ cells) do not cause GVHD in the B6(H-2b) →BALB/c (H-2d) fully MHC-mismatched allogeneic bone marrow transplant (BMT) model (27). In contrast, in the present work we demonstrate that highly purified CD8+CD62L+CD44+ TCM clearly induce GVHD, albeit less severe than that induced by TN. We did so in the same MHC-mismatched model used by Chen et al and in an MHC-matched, multiple minor histocompatibility antigen (miHA)-mismatched strain pairing. CD8+ TCM were also potent mediators of GVL.
Materials and Methods
Mice
Allogeneic bone marrow transplant (alloBMT) recipients and mice used to generate mCP-CML were 7–10 weeks of age. T cell and BM donors were between 8–16 weeks of age. B6 and BALB/c mice were obtained from the NCI (Frederick, MD). C3H.SW mice were obtained from the Jackson Labs (Bar Harbor, ME) or bred at Yale from breeding pairs purchased from the Jackson Labs.
Cell purifications
For GVHD experiments, CD8+ T cells were enriched from total spleen cells using negative selection with BioMag® magnetic beads (Qiagen, Hilden, Germany). Cells were incubated for 30 minutes on ice with the following antibody supernatants (all lab-grown): anti-CD4 (GK1.5), anti-B220 (RA3–6B2), anti-CD11b (M1/70), anti-FcR (24G.2). Cells were then washed in MACS buffer (PBS, 2mM EDTA, 0.5% BSA) and incubated with BioMag® goat anti-rat magnetic beads for 30 minutes on ice in a T75 or T125 flask, which was then placed next to a strong magnet. Cells not bound to magnetic particles were collected and were 60–70% CD8+. For further separations of TN and TCM, enriched CD8+ cells were incubated with anti-CD8-PE (53–7.6, BD Pharmingen), anti-CD62L-Alexa488 (Mel-14, lab-prepared), anti-CD44-APC (Pgp-1, BD Pharmingen) for 20 minutes on ice, washed and incubated with streptavidin-PE (BD Biosciences, San Jose, CA) for 15 minutes on ice, washed again and resuspended in PBS with 0.5% FBS. Cells were sorted into CD8+CD62L+CD44− TN and CD8+CD62L+CD44+ TCM using a FACS Aria (BD Biosciences). Bone marrow (BM) was depleted of T cells as previously described (28). For experiments wherein intracellular cytokine expression and the in vivo CTL assay were performed, CD8+ T cells were positively selected from splenocytes using anti-CD8α beads (Miltenyi) and the AutoMACs, stained with antibodies against CD8, CD62L and CD44 and sorted on the FACS Aria as described above. For analysis of cytokine production from freshly isolated TCM and TN, splenocytes were sorted using anti-CD8−Pacific Blue (clone TIB105, lab-prepared), anti-CD44−APC, and anti-CD62L−PE (BD Biosciences). B6 and BALB/c B cells were purified by depleting T cells from LN cells using anti-Thy1.2-biotin and streptavidin (SA)-microbeads (Miltenyi) and the AutoMACs.
GVHD transplant protocol
All transplants were performed according to protocols approved by the Yale University Institutional Animal Care and Use Committee. On day 0, B6 or BALB/c mice were irradiated (1000 cGy or 800cGy, respectively) and reconstituted with 7×106 T cell-depleted (TCD) C3H.SW or B6 BM, with no T cells, CD8+ TN, or CD8+ TCM (all cells given intravenously). Mice were weighed approximately every three days. GVHD in the B6→BALB/c model was nonlethal. Based on a large experience in the C3H.SW→B6 strain pairing (28–30), deaths prior to day 13 (which occurred equally in recipients of BM alone, TN and TCM) were attributed to the acute toxicity of transplant and not to GVHD and these events were censured in the survival analyses.
Histologic analysis
Tissues were fixed in 10% phosphate buffered formalin, paraffin-embedded, sectioned, and stained with hematoxylin and eosin. Slides were read by pathologists expert in skin (J.M.), liver and gastrointestinal disease (D.J.) without knowledge as to experimental group. Scoring was as described previously (28). Images of bowel and liver were obtained with an Olympus BX40 microscope (Olympus America, Lakewood, CO) using a 10× eyepiece and an 20× objective, with a QImaging QColor5 camera (Olympus) and QCapture software (Olympus). Images were processed with Adobe Photoshop 7.0 software (Adobe, San Jose, CA). Images of skin were obtained with an Olympus BX61 microscope (Olympus) using a 10× eyepiece, 20× objective, a SPOT RT Slider camera (Diagnostic Instruments, Sterling Heights, MI), SPOT software version 4.09 (Diagnostics Instruments) followed by processing with Adobe Photoshop 8.0.
Immunofluorescent staining
Tissues were fixed in 0.7% formaldehyde overnight followed by dehydration in 30% sucrose and freezing in Tissue-TeK OCT compound (Sakura Finetek; Torrance, California). 7 micron sections were incubated overnight at 4°C with DAPI and antibodies against CD8 (Alexa647; clone TIB105; lab-prepared). Sections were imaged with an Olympus BX40 microscope using a 10× eyepiece, 40× objective, a Scanalytics SPOT RT Slider camera (model 2.3.1, Diagnostic Instruments, Sterling Heights, MI) using SPOT software version 4.06 (Diagnostics Instruments). Pictures were reconstituted with Adobe Photoshop 8. To quantitate infiltration by CD8 cells, we counted CD8 cells in four 40× fields each from two mice that received only BM, 3 or 4 fields from two mice that received TN, and 3 or 4 fields from 2 mice that received TCM. We treated the number of CD8 cells in a field as an independent observation and compared numbers of cells using Mann-Whitney (Graphpad Prism).
Intracellular cytokine staining
Intracellular cytokine staining was performed as we have described (26). Briefly, splenocytes and LN cells (interscapular, axillary, cervical, mesenteric and inguinal) were harvested and cultured with phorbol myristic acid (PMA) and ionomycin for 5 hours; monensin was added for the final 2 hours. Prior to permeabilization, cells were incubated with ethidium monoazide (EMA) to allow exclusion of dead cells. Cells were stained with antibodies against CD45.1 (PE, BD Biosciences), CD8 (Pacific Blue, clone TIB105, lab prepared), permeabilized and then stained with antibodies against IFN-γ (APC; clone XMG1.2, BD Biosciences) and TNF-α (FITC, clone MP6-XT22, BD Biosciences) or isotype controls (for the anti-cytokine antibodies). Analysis was performed on an LSRII (BD Biosciences).
In vivo CTL assay
Equal numbers of LN-derived B cells from B6 and BALB/c mice were pooled and stained with carboxyfluoresceindiacetate succinimidyl ester (CFSE; Invitrogen, Carlsbad, CA). On day 7 post BMT, 107 B cells were injected into BALB/c recipients of TCD donor B6 BM, with no B6 T cells, or 106 sort-purified B6 TCM or TN.
Cytokine measurements
Serum cytokines were measured using the Bioplex mouse Th1/Th2 panel (Bio-Rad, Hercules, CA) and a BioPlex system.
Retrovirus production and progenitor infections
MSCV2.2 expressing the human bcr-abl p210 cDNA and a non-signaling truncated form of the human low affinity nerve growth factor receptor (NGFR) driven by an internal ribosome entry site (Mp210/NGFR) was a gift of Warren Pear. Retroviral supernatants were generated by transfection of Plat-E retrovirus packing cell line as described (28, 29, 31, 32). p210-infected bone marrow progenitors were generated as previously described (28, 29, 32). Briefly, B6 mice were injected on day −6 with 5mg 5-fluorouracil (5FU; Pharmacia & Upjohn, Kalamazoo Michigan). On day −2, BM cells were harvested and cultured in prestimulation media (DMEM, 15% FBS, IL-3 (6 ng/ml), IL-6 (10 ng/ml), and SCF (10 ng/ml) (all cytokines from Peprotech; Rocky Hill, New Jersey). On days –1 and 0 cells underwent “spin infection” with Mp210/NGFR retrovirus.
GVL transplant protocol
On day 0, host mice received 900cGy (450cGy × 2) and were reconstituted with 5×106 TCD C3H.SW BM with 7×105 B6 BM cells that underwent spin-infection with retroviral supernatant, with or without donor CD8+ TN or TCM. Cause of death was attributed to mCP-CML if mice had a high NGFR+ blood count on a recent analysis of peripheral blood (PB) and by spleen weight at necropsy.
Flow cytometry analysis in GVL experiments
Whole blood was stained with antibodies against Gr-1 (FITC; clone RB6–8C5, BD Pharmingen), TER-119 (PE; clone TER-119, BD Pharmingen Biosciences) and NGFR (Alexa647; clone 20.4; lab-prepared) followed by RBC lysis with ACK (Cambrex Bio Science, Walkersville, MD). Propidium iodide was added to exclude dead cells. BM and splenocytes were stained following ACK lysis. Cells were analyzed on a FACS Calibur (Beckton Dickinson Immunocytometry Systems, San Jose CA) and results analyzed with FlowJo (Treestar, Ashland, OR).
Statistical methods
Significance for differences in weight loss was calculated by an unpaired t-test evaluating measurements from all mice in a given group against all mice in a second group as we have previously described (22, 26, 28, 30). P-values for survival curve were calculated by log rank test. P-values for comparisons of histology scores and numbers of NGFR+ cells in blood were calculated by Mann-Whitney. P-values comparing numbers of TN or TCM derived cells and serum cytokine measurements were calculated using an unpaired t test (GraphPad Prism).
Results
CD8+ TCM induce GVHD in MHC-mismatched and MHC-matched alloBMT models
We first studied CD8+ TCM in the MHC-mismatched B6 (H-2b) → BALB/c (H-2d) model. Lethally irradiated BALB/c mice were reconstituted with TCD B6 BM with no T cells or with 1×106 CD8+CD62L+CD44− TN or CD8+CD62L+CD44+ TCM (Figure 1A). Beginning early post-BMT, CD8+ TN recipients lost weight relative to recipients of only TCD BM (Figure 1B) and this weight difference persisted until the experiment was concluded. CD8+ TCM recipients also lost weight relative to recipients of only TCD BM. However, at most timepoints past day 7, they had less weight loss than did recipients of CD8+ TN, and by the conclusion of the experiment, weight change was indistinguishable from recipients of only TCD BM. Thus, as measured by weight change, CD8+ TCM induced GVHD, but less than did CD8+ TN. 2/14 TN recipients died prior to day 8 and no deaths occurred in TCM recipients. Mice were sacrificed at approximately day +40 post transplant to harvest skin, small intestine, colon and liver for histologic GVHD. In both TN- and TCM-recipients GVHD was only present in the liver, manifest by periportal inflammatory infiltrates and bile duct damage (Figure 1C; representative pathology, Supplemental Figure 1) with a trend towards lower scores in TCM recipients (P=0.086; two-tailed).
Figure 1. CD8+ TCM induce milder but definite GVHD in the B6→BALB/c strain pairing.
A. Cell sorting. TN and TCM CD8 cells were purified and sorted from splenocytes as described in Methods. We gated on CD8+ cells (first panel), which were sorted into TN and TCM based on the expression of CD62L and CD44. B and C. GVHD in B6→BALB/c model (data combined from 2 experiments). BALB/c mice were lethally irradiated and reconstituted with TCD B6 BM, with no T cells or 1×106 CD8+ TN or TCM. B. Weight change. P<0.005 at days 5, 8, 12 and 14 comparing recipients of BM versus CD8+ TN or TCM; P=0.013 comparing BM alone versus TN on day 20. P<0.01 at day 20, 23, 27, 30, 34, 37 and 40 comparing recipients of CD8+ TN versus CD8+ TCM. C. Liver pathology. Each symbol represents a score from an individual mouse; horizontal lines are mean scores. P=0.0018 comparing BM and CD8+ TN recipients; P=0.0085 comparing BM and CD8+ TCM recipients. P=0.086 comparing CD8+ TN versus CD8+ TCM. D and E. BALB/c mice were irradiated and reconstituted with TCD B6 BM, with no T cells, 1×106 TN, 1×106 TCM or 2×104 TN (TN control) B6 CD8 cells. D. Weight change. E. Liver pathology scores. P<0.005 for recipients of TN or TCM versus TN control. P value is not significant between BM and TN control recipients.
Sorted CD8+ TCM had between 1% and 2% contaminating CD8+ TN. We therefore in the second of the two experiments from which data was combined in Figures 1B and 1C, included a group that received 2×104 CD8+ TN (TN control), the number of TN in the sorted CD8+ TCM. 1×106 CD8+ TCM and CD8+ TN induced both weight loss and hepatic GVHD, whereas 2×104 CD8+ TN induced neither (Figure 1, D and E). Thus the GVHD induced by CD8+ TCM was not due to contaminating CD8+ TN.
Next we determined whether sorted CD8+ TCM mediate GVHD in the C3H.SW(H-2b)→ B6 (H-2b) MHC-matched, multiple miHA-mismatched strain pairing. Lethally irradiated B6 mice were reconstituted with C3H.SW TCD BM with no T cells or with 1.5×106 CD8+ TN or TCM. Recipients of only TCD C3H.SW BM did not develop GVHD whereas most CD8+ TN recipients developed clinical GVHD manifest by death (Figure 2A; 40% by day 43; P=0.0029 comparing BM and TN), weight loss (Figure 2B), diarrhea, ruffled fur and skin lesions (not shown). In contrast, only 1/13 CD8+ TCM recipients died by day 43 (P=0.2138 comparing BM and TCM; P=0.054 comparing TN and TCM). Weight loss in TCM-recipients was less than in recipients of TN and not statistically different than recipients of only donor BM, though there was a trend towards lower weights (as compared to BM controls) at late time-points. Tissues were harvested for histologic analysis on day +43 post BMT. TCM caused statistically significant pathologic GVHD in the skin, liver and small intestine (Figure 2C; representative histology in Supplemental Figure 1), whereas TN induced significant GVHD in these tissues and in the colon (Figure 2C). Although TCM did not induce GVHD in the colon (relative to BM alone controls), scores in comparison to TN recipients did not reach significance (P=0.159). In the liver, aside from 3 mice with high scores, the majority of mice had relatively mild liver GVHD. Nonetheless, even if the scores from these mice are excluded or assigned a value of 5, the P value comparing liver GVHD in BM alone controls and TCM recipients is still significant (P<=0.021), though this would render disease in TN recipients more severe than in TCM recipients (P<=0.043). Thus, CD8+ TCM induce GVHD in the C3H.SW→B6 strain pairing. However, GVHD was clinically less severe than that induced by TN and this difference may at least in-part be explained by less colon and liver GVHD in TCM recipients.
Figure 2. CD8+ TCM induce milder but definite GVHD in C3H.SW→B6 strain pairing.
Data combined from 3 experiments. B6 mice were lethally irradiated and reconstituted with TCD C3H.SW BM, with no T cells, 1.5×106 CD8+ TN or TCM. A. Survival. P=0.0029 comparing BM and TN; P=0.2138 comparing BM and TCM; P=0.054 comparing TN and TCM. B. Weight change. P<0.001 at days 26, 30, 34, 37 and 43 comparing recipients of BM only versus CD8+ TN; P<0.05 at days 26 and 43 comparing recipients of TN versus TCM. P values not significant at any time-point comparing BM alone versus TCM. C. Pathology scores. Each symbol represents a score from an individual mouse; horizontal lines are mean scores. P values are noted below each panel.
To further investigate the nature of GVHD in B6 recipients of C3H.SW TCM we performed immunofluorescence staining on small bowel specimens to identify infiltrating CD8+ T cells. CD8+ T cells invaded the mucosa with penetration into villi in both TCM and TN recipients while such cells were rare in recipients of only TCD C3H.SW BM (Figure 3A, representative images; Figure 3B, number of CD8 cells/40× field). CD8 cells were more frequent in TCM and TN recipients than in recipients of only BM (P=0.003) whereas counts in TCM and TN recipients were similar (P=0.8). Thus TN- and TCM-derived effectors shared the ability to cause infiltrative GVHD of the small intestine.
Figure 3. CD8+ TN and TCM infiltrate small bowel in the C3H.SW→B6 strain pairing.
Frozen sections of small bowel from recipients of only TCD BM and TN or TCM recipients with confirmed histologic GVHD (by blinded scoring) were stained with anti-CD8 (Alexa647; rendered in green). DAPI-stained nuclei are rendered in blue. Shown in A are representative sections from two recipients of TN, TCM or only TCD BM. In TN and TCM recipients, CD8 cells percolate through the bowel and invade crypts, characteristic of GVHD. B, quantitation of the number of CD8 cells. CD8 cells were counted in 3–4 40× fields from two mice from each group. Each circle represents the number of CD8 cells in an individual field; horizontal lines are means. P=0.003 comparing BM alone versus TN or TCM; P=0.8 comparing TN and TCM.
CD8+ TCM mediate GVL
To study the efficacy of CD8+ TCM in mediating GVL, we used a mouse model of chronic phase CML (mCP-CML) created via retroviral transduction of BM cells with the human bcr-abl fusion cDNA (p210) (28, 29). The construct also expresses a nonsignaling form of NGFR, which allows detection of mCP-CML cells by flow cytometry. mCP-CML is characterized by a high white blood cell count and splenomegaly, with hematopoiesis dominated by maturing myeloid cells (32, 33). In the C3H.SW→B6 strain pairing GVL is directed towards miHAs and not against p210 and NGFR (29, 34). B6 recipients were lethally irradiated and reconstituted with TCD C3H.SW BM, p210-infected B6 BM and no C3H.SW T cells or 3× 105 CD8+ TN or TCM. We chose a low dose of donor T cells that mostly prolong survival rather than curing 100% of mice, thereby allowing the detection of minor differences in the abilities of TN and TCM to induce GVL that could be masked by higher T cell doses. All mice that did not receive donor CD8+ T cells died from mCP-CML by day 22. In contrast, recipients of either CD8+ TN or TCM had prolonged survival with 1/10 in each group surviving to the end of the experiment at day 75 (Figure 4A; all deaths were from mCP-CML). TCM-mediated GVL was not due to contaminating TN as recipients of 7.5×103 CD8+ TN (TN control), the number of TN in the sorted CD8+ TCM cells, died from mCP-CML with similar kinetics as did mice that received no donor T cells (Figure 4A). We serially analyzed peripheral blood using NGFR expression to identify mCP-CML cells (Figure 4B, representative flow cytometry). Overall TN and TCM recipients had comparable numbers of NGFR+ cells (Figure 4C) with TCM-recipients having fewer NGFR+ cells on day +11 (P=0.0039), more on day +18 (P=0.0185), without statistically significant differences at the remaining time-points. Similar results were observed when 1×105 TN and TCM were used to induce GVL (not shown). Thus, CD8+ TN and TCM have comparable capacities to induce GVL.
Figure 4. CD8+ TCM mediate GVL against mCP-CML.
B6 mice were irradiated and reconstituted with TCD C3H.SW BM, B6 mCP-CML with no T cells, 3×105 CD8+ TN, 3×105 CD8+ TCM, or 7.5×103 CD8+ TN (TN control) C3H.SW CD8 cells. A. Survival data. P<0.001 for recipients of CD8+ TN or CD8+ TCM versus only BM. B. Representative serial flow cytometry of peripheral blood. Each column is from an individual mouse; two representative mice are shown for TN and TCM recipients. C. Numbers of NGFR+ cells in the peripheral blood of transplanted mice at different time points. Each symbol represents an individual animal; solid lines are mean values.
TCM and TN expansion, cytokine production and cytolytic activity post alloBMT
To better understand how donor TCM and TN expand and mature into effectors after transfer, we analyzed progeny of TCM and TN 7 days after BMT in the B6→BALB/c model wherein GVHD was induced by 106 CD8+ TN or TCM. We used CD45.1+ and CD45.2+ B6 mice as T cell and BM donors, respectively, so as to be able to clearly identify infused TN and TCM (BALB/c mice are CD45.2). As controls we performed syngeneic B6→B6 transplants. Both BALB/c TCM- and TN-recipients had approximately 4×106 and 4×105 CD45.1+CD8+ cells in spleen and lymph nodes, respectively (Figure 5, C and D; P>0.6 comparing TN and TCM recipients). This expansion was dramatically greater than was seen in B6 recipients of TCM or TN wherein spleens and LNs contained 20–30-fold and 10-fold fewer CD8+CD45.1+ cells, respectively (P<0.0015 comparing BALB/c versus B6 recipients of TN or TCM). Although it is unlikely that in BALB/c recipients all progeny of TCM and TN at day +7 are alloreactive, the difference in expansion of infused TCM and TN in BALB/c relative to B6 hosts is attributable to the presence of and effects from alloreactive T cells, and by this measure TN and TCM behaved similarly. Interestingly, relative to TN-recipients, a greater number of TCM-derived cells were found in B6 spleens (P=0.026) but not in LN. Decreased expression of CD8 is a marker for T cell activation. Although sorted CD8+ TCM and TN had a similar fluorescent intensity prior to transfer, in syngeneic transplants TCM progeny had a lower mean geometric mean fluorescent intensity than did TN (TCM: 11870 +/− 520; TN: 26110 +/−2051; P=0.0025). CD8 expression on both TN and TCM was dramatically downregulated in allogeneic recipients (TCM: 4388 +/−211; TN: 4642 +/−186; P=0.42; comparing TN to TCM in BALB/c recipients; P<= 0.0005 comparing TN or TCM in BALB/c versus B6 recipients).
Figure 5. Both TN and TCM expand in syngeneic and allogeneic recipients and produce IFN-γ.
BALB/c (CD45.2) and B6 (CD45.2) mice were irradiated and reconstituted with TCD B6 CD45.2 BM and 106 CD8+CD45.1+ TN or TCM. Seven days post BMT, spleen and LN cells were stimulated with PMA and ionomycin and then stained for CD45.1, CD8, IFN-γ and TNF-α. Positive gates for IFN-γ were based on isotype staining (not shown). A and B, representative flow cytometry of CD8+CD45.1+ splenocytes and LN cells, respectively. The percentage of IFN-γ+ and IFN-γ very bright cells are shown in the upper left and upper right corners, respectively. Total numbers of CD45.1+CD8+, CD45.1+CD8+IFN-γ+ or IFN-γ-bright and the percent of CD8+CD45.1+ cells that are IFN-γ+ is shown for spleen (C) and LN (D). Each circle is the value from an individual mouse; horizontal lines are mean values. E, IFN-γ expression after PMA/ionomycin treatment of freshly isolated TCM (right panel) and TN (left panel).
To analyze differentiation into effectors, spleen and LN cells from transplant recipients (isolated on Day+7) were stimulated with PMA and ionomycin and analyzed by intracellular cytokine staining for the presence of IFN-γ and TNF-α. We observed little, if any, TNF-α production (Figure 5A, representative staining). However, approximately 40–50% of CD8+CD45.1+ spleen cells in BALB/c recipients of TN or TCM produced IFN-γ, with comparable overall numbers of both IFN-γ+ and IFN-γ-very bright cells in TN and TCM recipients (Figure 5A, representative staining; Figure 5C, quantitation; P>0.29). The percentage of TN or TCM progeny that were IFN-γ+ in LN was similar to that in spleen (Figure 5B, representative staining; Figure 5D, quantitation); however, a smaller percentage were IFN-γ very bright (P<0.02 comparing TN or TCM in LN versus those in spleen). Syngeneic recipients of TCM had more splenic CD8+CD45.1+IFN-γ+ cells than did TN recipients, which paralleled the difference in overall numbers of donor-derived CD8 cells (P=0.028). We were also interested in how well freshly isolated B6 CD8+ TCM and TN produced IFN-γ after PMA/ionomycin stimulation (Figure 5E). Only 9% of TN produced IFN-γ, and only weakly. In contrast, 70% of TCM produced IFN-γ, with a geometric mean expression nearly 6-fold higher than in TN. Nearly 25% of TCM brightly expressed IFN-γ as compared to no IFN-γ-bright TN. In sum, after transfer to BALB/c recipients, TCM and TN similarly differentiated into IFN-γ-producing cells, although TCM may have already been polarized to do so.
We also analyzed serum samples taken on day +7 from these transplant recipients for cytokines using the Bio-Plex system. We analyzed levels of IFN-γ, IL-2, TNF-α (Figure 6), IL-12 and GMCSF (not shown). Of these, only IFN-γ was elevated in allogeneic recipients relative to syngeneic recipients, (P<0.02 comparing TN or TCM in B6→BALB/c versus B6→B6 recipients). There was no difference in IFN-γ levels in allogeneic TCM versus TN recipients (P=0.41).
Figure 6. Allogeneic TCM and TN induce serum IFN-γ.
Serum was harvested from B6→B6 and B6→BALB/c recipients of B6 TN or TCM on day +7. Cytokine concentrations were determined by Bio Plex beads. IFN-γ but not IL-2 or TNF-α were elevated in allogeneic TCM or TN recipients. Each symbol is a measurement from an individual mouse; horizontal lines are means.
To compare cytolytic function of transferred TN and TCM, we injected 5×106 CFSE-labeled B6 and 5×106 CFSE-labeled BALB/c B cells into BALB/c mice that had been transplanted 7 days prior with TCD B6 BM, with no T cells or with 106 CD8+ TN or TCM. Twenty hours later, mice were sacrificed and the survival of CFSE+B220+ B6 and BALB/c cells in spleen and LN was assessed by flow cytometry. In recipients of only TCD BM, the ratio of infused B6 and BALB/c B cells was nearly 1:1 (Figure 7). In contrast, in both TN and TCM recipients, greater than 94% of CFSE+B220+ cells were H-2Kb+ and therefore B6 in origin. Thus, both TN and TCM mediated potent killing of allogeneic BALB/c B cells, consistent with their GVL activity.
Figure 7. Allogeneic BALB/c B cells are preferentially killed in vivo in recipients of B6 TCM or TN 7 days post transplant.
Representative flow cytometry of splenocytes (SPL) and lymph node cells (LN). Plots are gated on B220+ cells. Note that greater than 94% of CFSE+B220+ cells are H-2Kb+ and therefore B6 in recipients of TN or TCM.
Discussion
Prior studies have shown that TEM not specifically primed to alloantigens are greatly impaired in their ability to induce GVHD (22–27, 35, 36). The capacity of TCM to do so has been less well studied, and one group recently reported in a single MHC-mismatched model that TCM do not cause GVHD (27). In contrast to that study, in the present work we clearly show in MHC-matched and MHC-mismatched strain pairs that CD8+ TCM not specifically primed to alloantigens do cause clinical and pathologic GVHD. However, TCM were less potent as measured by weight loss, death, and by less pathologic colonic GVHD in the C3H.SW→B6 strain pairing. There was also a suggestion that overall, TCM-induced less severe liver GVHD in both models. Thus our in-depth studies show that TCM are capable of inducing GVHD and that they differ substantially from TEM, but more subtly from TN.
In contrast to their abilities to induce GVHD, CD8+ TCM and TN were comparable mediators of GVL as measured both by survival and by the numbers of NGFR+ mCP-CML cells in peripheral blood at several time points. For GVL experiments we employed a T cell dose well below that which cures 100% of mice so as to be able to detect small differences in T cell potency, making it unlikely that we missed a major difference. These data are consistent with prior work with TCR-transgenic TCM in an antitumor model (17, 19). Thus the selective infusion of TCM may be able to mediate GVL with less GVHD, though the reduction in GVHD may only be modest.
In our studies, TCM were not generated specifically against alloantigens. Rather they likely differentiated into TCM after reacting against allergens or commensual and pathogenic organisms that can infect laboratory mice (37–42). Alternatively, these TCM phenotype cells could be the product of lymphopenia-induced proliferation which occurs early in the development of the immune system (43–46). Consistent with having been previously activated, 70% of freshly isolated CD8+ TCM produced IFN-γ after PMA/ionomycin stimulation. Regardless of their precise activation history, the TCM used in our studies clearly responded against both allogeneic MHC-peptide and syngeneic-MHC-miHA peptide complexes. Such cross-reactivity, or heterologous immunity, has been well-demonstrated in T cells reactive against both allogeneic MHC (47–62) and against viral peptides on a shared MHC (61, 63–73), analogous to our results in the B6→BALB/c and C3H.SW→B6 models, respectively. Alloreactive memory cells are thought to be barriers to successful solid organ transplantation (48, 51–53, 55, 74), and the present data that TCM mediate both GVHD and GVL are consistent with these studies.
B6 CD8+ TCM and TN similarly expanded by day +7 in allogeneic recipients, and expansion of both was 20–30-fold greater than in syngeneic recipients. This difference highlights the alloreactivity in both populations. It is surprising that even though TCM and TN differ in TCR repertoire and in their prior activation history, that similar numbers accumulated in spleen and LN. In contrast, in syngeneic transplants, TCM expanded to larger numbers. These data suggest that there may be T cell intrinsic and/or extrinsic factors that restrict T cell expansion in allogeneic recipients, such as limitation on the amount of antigen (in this case in the form of recipient antigen presenting cells) or cytokines. This picture parallels observations in other models wherein the peak of antigen-driven T cell expansion is not directly proportional to the number of precursor cells that initially are activated (75, 76).
Both TCM and TN progeny expressed effector functions in vitro and in vivo. Similar fractions of TN or TCM-derived cells produced IFN-γ in both syngeneic and allogeneic recipients, though a greater percentage of both TN and TCM were IFN-γ-bright in allogeneic recipients. Although we cannot determine whether IFN-γ-producing progeny of TCM were derived from cells that had previously been polarized to produce IFN-γ, given that 70% of freshly isolated TCM produce IFN-γ, it is reasonable to suggest that the precursors of many of the TCM progeny were already polarized. In contrast, most TN were likely polarized after transfer in both syngeneic and allogeneic recipients. Allogeneic TN or TCM recipients also had elevated serum IFN-γ levels which roughly paralleled the numbers of IFN-γ producing cells.
We also compared CTL activity of TN and TCM progeny against allogeneic targets in vivo, and both specifically killed BALB/c B cells relative to syngeneic B6 B cells. These data do not address the frequency of TN or TCM-derived CTL that recognize allogeneic targets or their specific potencies on a per cell basis; however they clearly demonstrate that both TCM and TN differentiate into CTLs as indicated in the GVL experiments.
Our results differ from those of Chen and colleagues who reported that a mix of CD4+ and CD8+ TCM induced neither clinical nor histologic GVHD in the B6→BALB/c strain pairing, which we also used. In that study, TCM were identified as being CD62L+CD44+CD45RB+ whereas we did not stain for CD45RB. In our hands and in their published data, CD8+CD62L+CD44+ cells are all CD45RB+ and therefore the inclusion of CD45RB should not have resulted in their sorting a population distinct from that in our work. In their experiments, 106 total TCM were transferred, of which approximately half were CD8+. Thus it is possible that GVHD was not induced because fewer CD8+ TCM were transferred. If so then 5×105 CD4+ TCM must neither induce GVHD nor promote the activation of alloreactive CD8+ TCM. The failure of a mix of CD4+ and CD8+ TCM to induce GVHD was not due to CD62L+CD44+CD4+CD25+ regulatory T cells as in their studies, when these cells were depleted, TCM still did not induce GVHD. CD4+ TCM may in some other way reduce the capacity of CD8+ TCM to cause GVHD, though this is not generally the case with unfractionated CD4 and CD8 cells in GVHD models (77).
We point out that Chen et al reported a negative result, whereas we report a positive one—that is TCM induced both GVHD and GVL. Therefore one must consider other possible reasons that Chen et al failed to make this observation. It is conceivable that the cell sorting procedure these investigators employed sufficiently reduced the viability, survival after transfer, or functionality of TCM so as to result in an underestimation of their GVHD-inducing potential. In their experiments, sorted TCM contained 3% and therefore 30,000 TN whereas 10,000 unfractionated T cells (of which only ~50% were naïve) that did not undergo cell-sorting, induced death and weight loss in BALB/c recipients. Therefore if the majority of sorted cells were viable, one would have anticipated that some GVHD would have been seen in recipients of 30,000 contaminating TN, unless this was suppressed by the majority TCM. It is also possible that antibody bound to CD45RB altered the survival or function of the sorted TCM (78). Regardless of why our results differ, our data are a critical contribution in showing with multiple models, that CD8+ TCM indeed do cause GVHD.
As to why CD8+ TCM have a greater capacity to induce GVHD than do CD8+ TEM, we see several hypotheses. One possibility is that TCM may have a broader T cell receptor (TCR) repertoire that includes more receptors that recognize alloantigens. In humans, TEM and TCM have distinct but similarly complex repertoires (79). However, it is difficult to extrapolate this data to laboratory mice in which the repertoire of these subsets is less likely to have been shaped by constant exposure to pathogens. TCM may be more efficiently activated after transfer due to better access to LN and Peyer’s patches, owing to their expression of CD62L and CCR7. Such a mechanism has been postulated to at least in-part explain why in vitro generated CD8+ TCR-transgenic TCM were more potent in an anti-tumor model than were TEM (17). However, it was recently shown that TN induce robust GVHD in mice that nearly completely lack LNs and Peyer’s patches (PP) (80, 81). Nonetheless it remains to be tested whether TCM are more reliant on priming in those sites than are TN.
Another possibility is that TCM are more capable than TEM of undergoing clonal expansion. TCM have a greater in vitro proliferative capacity (reviewed in (10)). In vivo, adoptively transferred TCR-transgenic TCM provide greater protection from LCMV infection than do adoptively transferred TEM, and this was shown to be due to a greater proliferative capacity of TCM (11). The relative abilities of CD8+ TEM and TCM to protect from pathogens is model-dependent and features other than proliferative capacity play a role; nonetheless, overall TCM seem to have a greater ability to undergo clonal expansion (reviewed in (20)). These types of quantititative analyses are not possible in our studies, which used naturally occurring polyclonal TCM, due to the inability to identify miHA-stimulated T cells and distinguish them from those that underwent lymphopenia-induced proliferation. In alloBMT models, CD4+ TEM do not accumulate as well as TN (23, 26), but again T cells activated by alloantigen could not be distinguished from those that underwent lymphopenia-induced proliferation.
A related question is why TCM induced milder clinical and subtly less pathologic GVHD than did TN. Both TN and TCM have ready access to LN and PPs due to their expression of CD62L and CCR7. Also, our data demonstrated that both TN- and TCM-derived effectors were able to infiltrate target tissues (Figure 3). However, TCM may more readily migrate directly into tissues after infusion, thereby effectively reducing the number of cells available for activation in secondary lymphoid tissues. For example, TCM more efficiently enter BM spaces (82). Nonetheless, by day 7 there were comparable numbers of TCM and TN-derived cells in both spleen and LN. TCM may have a less alloreactive TCR repertoire than do TN, and again this could be more applicable in humans wherein the TCR repertoire of memory phenotype cells is more likely to have been shaped and narrowed by pathogens (83–86). In MHC-matched, multiple miHA-disparate transplants, not only could the frequency of alloreactive T cells be reduced overall in TCM, but the specific miHAs targeted could be different as the TCR repertoire can impact on the choice of immunodominant antigens (87–91). Thus TCM could have chosen a different set or hierarchy of immunodominant antigens, which is known to be a property of T cell responses to miHAs (92–94).
There could also be differences in other intrinsic properties of the progeny of activated TN and TCM, such as the types of cytokines elaborated, expression of cytotoxic granules, TNF family ligands and resistance to activation-induced cell death. At a minimum we know that in the B6→BALB/c model, TN and TCM-derived cells both produced IFN-γ and had cytolytic function in vivo. However, given that such a high frequency of freshly isolated TCM produced IFN-γ, it is likely that a substantial fraction of alloreactive TCM were already programmed. It is possible that they differentiated into effectors less capable of causing disease than did TN only activated in BMT recipients. Investigating potential differences between TCM and TN will require comparing in GVHD models TN genetically deficient in candidate molecules by which they may differ from TCM (reviewed in (95–97)) to intact TCM; given the likelihood that multiple factors contribute and the relatively mild quantitative differences in GVHD induction, such studies are not likely to be informative.
A complete understanding of the relative potencies and mechanisms of action of TCM, TEM and TN in GVHD and GVL models will require experimental systems wherein donor TN, TCM and TEM have equivalent and defined TCR repertoires, which would help to control at least some of these variables. Nonetheless, with available systems, it is important conceptually, mechanistically and clinically to know that CD8+ TCM can cause both GVHD and GVL, in contrast to what was previously thought (27).
Supplementary Material
Acknowledgements
We thank the Yale Animal Resources Center for expert animal care. We also thank Srividhya Venkatesan and Hung Sheng Tan for dedicated technical assistance.
This work was supported by NIH grant R01-CA96943 and R01-HL 066279. W.D.S is a recipient of a Clinical Scholar award from the Leukemia and Lymphoma Society. H.Z. was a recipient of Christian Jacobsen Postdoctoral Fellowship Award from The Marrow Foundation and the National Marrow Donor Program.
Footnotes
Disclosures
The authors have no conflicting financial interests.
References
- 1.Mackall CL, and Gress RE 1997. Pathways of T-cell regeneration in mice and humans: implications for bone marrow transplantation and immunotherapy. Immunol Rev 157:61–72. [DOI] [PubMed] [Google Scholar]
- 2.Collins RH Jr., Shpilberg O, Drobyski WR, Porter DL, Giralt S, Champlin R, Goodman SA, Wolff SN, Hu W, Verfaillie C, List A, Dalton W, Ognoskie N, Chetrit A, Antin JH, and Nemunaitis J 1997. Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation. J Clin Oncol 15:433–444. [DOI] [PubMed] [Google Scholar]
- 3.Kolb HJ, Schattenberg A, Goldman JM, Hertenstein B, Jacobsen N, Arcese W, Ljungman P, Ferrant A, Verdonck L, Niederwieser D, and et al. 1995. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia. Blood 86:2041–2050. [PubMed] [Google Scholar]
- 4.Porter DL, Collins RH Jr., Shpilberg O, Drobyski WR, Connors JM, Sproles A, and Antin JH 1999. Long-term follow-up of patients who achieved complete remission after donor leukocyte infusions. Biol Blood Marrow Transplant 5:253–261. [DOI] [PubMed] [Google Scholar]
- 5.Horowitz MM, Gale RP, Sondel PM, Goldman JM, Kersey J, Kolb HJ, Rimm AA, Ringden O, Rozman C, Speck B, and et al. 1990. Graft-versus-leukemia reactions after bone marrow transplantation. Blood 75:555–562. [PubMed] [Google Scholar]
- 6.Sprent J, and Surh CD 2001. Generation and maintenance of memory T cells. Curr Opin Immunol 13:248–254. [DOI] [PubMed] [Google Scholar]
- 7.Berard M, and Tough DF 2002. Qualitative differences between naive and memory T cells. Immunology 106:127–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sallusto F, Lenig D, Forster R, Lipp M, and Lanzavecchia A 1999. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions [see comments]. Nature 401:708–712. [DOI] [PubMed] [Google Scholar]
- 9.Sallusto F, Mackay CR, and Lanzavecchia A 2000. The role of chemokine receptors in primary, effector, and memory immune responses. Annu Rev Immunol 18:593–620. [DOI] [PubMed] [Google Scholar]
- 10.Sallusto F, Geginat J, and Lanzavecchia A 2004. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol 22:745–763. [DOI] [PubMed] [Google Scholar]
- 11.Wherry EJ, Teichgraber V, Becker TC, Masopust D, Kaech SM, Antia R, von Andrian UH, and Ahmed R 2003. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat Immunol 4:225–234. [DOI] [PubMed] [Google Scholar]
- 12.Kalia V, Sarkar S, Gourley TS, Rouse BT, and Ahmed R 2006. Differentiation of memory B and T cells. Curr Opin Immunol 18:255–264. [DOI] [PubMed] [Google Scholar]
- 13.Masopust D, Vezys V, Wherry EJ, and Ahmed R 2007. A brief history of CD8 T cells. Eur J Immunol 37 Suppl 1:S103–110. [DOI] [PubMed] [Google Scholar]
- 14.Weninger W, Crowley MA, Manjunath N, and von Andrian UH 2001. Migratory properties of naive, effector, and memory CD8(+) T cells. J Exp Med 194:953–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bouneaud C, Garcia Z, Kourilsky P, and Pannetier C 2005. Lineage relationships, homeostasis, and recall capacities of central- and effector-memory CD8 T cells in vivo. J Exp Med 201:579–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zaph C, Uzonna J, Beverley SM, and Scott P 2004. Central memory T cells mediate long-term immunity to Leishmania major in the absence of persistent parasites. Nat Med 10:1104–1110. [DOI] [PubMed] [Google Scholar]
- 17.Klebanoff CA, Gattinoni L, Torabi-Parizi P, Kerstann K, Cardones AR, Finkelstein SE, Palmer DC, Antony PA, Hwang ST, Rosenberg SA, Waldmann TA, and Restifo NP 2005. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci U S A 102:9571–9576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, and Riddell SR 2008. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest 118:294–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hinrichs CS, Gattinoni L, and Restifo NP 2006. Programming CD8+ T cells for effective immunotherapy. Curr Opin Immunol 18:363–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lefrancois L 2006. Development, trafficking, and function of memory T-cell subsets. Immunol Rev 211:93–103. [DOI] [PubMed] [Google Scholar]
- 21.Roberts AD, Ely KH, and Woodland DL 2005. Differential contributions of central and effector memory T cells to recall responses. J Exp Med 202:123–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Anderson BE, McNiff J, Yan J, Doyle H, Mamula M, Shlomchik MJ, and Shlomchik WD 2003. Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest 112:101–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Beilhack A, Schulz S, Baker J, Beilhack GF, Wieland CB, Herman EI, Baker EM, Cao YA, Contag CH, and Negrin RS 2005. In vivo analyses of early events in acute graft-versus-host disease reveal sequential infiltration of T-cell subsets. Blood 106:1113–1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Chen BJ, Cui X, Sempowski GD, Liu C, and Chao NJ 2004. Transfer of allogeneic CD62L- memory T cells without graft-versus-host disease. Blood 103:1534–1541. [DOI] [PubMed] [Google Scholar]
- 25.Zhang Y, Joe G, Zhu J, Carroll R, Levine B, Hexner E, June C, and Emerson SG 2004. Dendritic cell-activated CD44hiCD8+ T cells are defective in mediating acute graft-versus-host disease but retain graft-versus-leukemia activity. Blood 103:3970–3978. [DOI] [PubMed] [Google Scholar]
- 26.Zheng H, Matte-Martone C, Li H, Anderson BE, Venketesan S, Sheng Tan H, Jain D, McNiff J, and Shlomchik WD 2008. Effector memory CD4+ T cells mediate graft-versus-leukemia without inducing graft-versus-host disease. Blood 111:2476–2484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chen BJ, Deoliveira D, Cui X, Le NT, Son J, Whitesides JF, and Chao NJ 2007. Inability of memory T cells to induce graft-versus-host disease is a result of an abortive alloresponse. Blood 109:3115–3123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Matte CC, Liu J, Cormier J, Anderson BE, Athanasiadis I, Jain D, McNiff J, and Shlomchik WD 2004. Donor APCs are required for maximal GVHD but not for GVL. Nat Med 10:987–992. [DOI] [PubMed] [Google Scholar]
- 29.Matte CC, Cormier J, Anderson BE, Athanasiadis I, Liu J, Emerson SG, Pear W, and Shlomchik WD 2004. Graft-versus-leukemia in a retrovirally induced murine CML model: mechanisms of T-cell killing. Blood 103:4353–4361. [DOI] [PubMed] [Google Scholar]
- 30.Shlomchik WD, Couzens MS, Tang CB, McNiff J, Robert ME, Liu J, Shlomchik MJ, and Emerson SG 1999. Prevention of graft versus host disease by inactivation of host antigen- presenting cells. Science 285:412–415. [DOI] [PubMed] [Google Scholar]
- 31.Morita S, Kojima T, and Kitamura T 2000. Plat-E: an efficient and stable system for transient packaging of retroviruses. Gene Ther 7:1063–1066. [DOI] [PubMed] [Google Scholar]
- 32.Pear WS, Miller JP, Xu L, Pui JC, Soffer B, Quackenbush RC, Pendergast AM, Bronson R, Aster JC, Scott ML, and Baltimore D 1998. Efficient and rapid induction of a chronic myelogenous leukemia-like myeloproliferative disease in mice receiving P210 bcr/abl-transduced bone marrow. Blood 92:3780–3792. [PubMed] [Google Scholar]
- 33.Li S, Ilaria RL Jr., Million RP, Daley GQ, and Van Etten RA 1999. The P190, P210, and P230 forms of the BCR/ABL oncogene induce a similar chronic myeloid leukemia-like syndrome in mice but have different lymphoid leukemogenic activity. J Exp Med 189:1399–1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Shlomchik WD, and Matte-Martone CC 2006. Leukemia-specific antigens alone are insufficient for GVL in MHC-matched allogeneic stem cell transplantation: An essential role for minor H antigens. Blood 108:59A–60A. [Google Scholar]
- 35.Dutt S, Ermann J, Tseng D, Liu YP, George TI, Fathman CG, and Strober S 2005. L-selectin and beta7 integrin on donor CD4 T cells are required for the early migration to host mesenteric lymph nodes and acute colitis of graft-versus-host disease. Blood 106:4009–4015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Dutt S, Tseng D, Ermann J, George TI, Liu YP, Davis CR, Fathman CG, and Strober S 2007. Naive and memory T cells induce different types of graft-versus-host disease. J Immunol 179:6547–6554. [DOI] [PubMed] [Google Scholar]
- 37.Fox JG 2002. Laboratory animal medicine. Academic Press, Amsterdam; New York. [Google Scholar]
- 38.Fox JG 2007. The mouse in biomedical research. Elsevier, AP, Amsterdam; Boston. [Google Scholar]
- 39.Foltz CJ, Fox JG, Cahill R, Murphy JC, Yan L, Shames B, and Schauer DB 1998. Spontaneous inflammatory bowel disease in multiple mutant mouse lines: association with colonization by Helicobacter hepaticus. Helicobacter 3:69–78. [DOI] [PubMed] [Google Scholar]
- 40.Fox JG, Yan LL, Dewhirst FE, Paster BJ, Shames B, Murphy JC, Hayward A, Belcher JC, and Mendes EN 1995. Helicobacter bilis sp. nov., a novel Helicobacter species isolated from bile, livers, and intestines of aged, inbred mice. Journal of clinical microbiology 33:445–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pappo J, Torrey D, Castriotta L, Savinainen A, Kabok Z, and Ibraghimov A 1999. Helicobacter pylori infection in immunized mice lacking major histocompatibility complex class I and class II functions. Infect. Immun 67:337–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Shames B, Fox JG, Dewhirst F, Yan L, Shen Z, and Taylor NS 1995. Identification of widespread Helicobacter hepaticus infection in feces in commercial mouse colonies by culture and PCR assay. Journal of clinical microbiology 33:2968–2972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Min B, Foucras G, Meier-Schellersheim M, and Paul WE 2004. Spontaneous proliferation, a response of naive CD4 T cells determined by the diversity of the memory cell repertoire. Proc Natl Acad Sci U S A 101:3874–3879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Min B, Yamane H, Hu-Li J, and Paul WE 2005. Spontaneous and homeostatic proliferation of CD4 T cells are regulated by different mechanisms. J Immunol 174:6039–6044. [DOI] [PubMed] [Google Scholar]
- 45.Surh CD, and Sprent J 2006. On the TRAIL of homeostatic memory T cells. Nat Immunol 7:439–441. [DOI] [PubMed] [Google Scholar]
- 46.Hamilton SE, Wolkers MC, Schoenberger SP, and Jameson SC 2006. The generation of protective memory-like CD8+ T cells during homeostatic proliferation requires CD4+ T cells. Nat Immunol 7:475–481. [DOI] [PubMed] [Google Scholar]
- 47.Lombardi G, Sidhu S, Daly M, Batchelor JR, Makgoba W, and Lechler RI 1990. Are primary alloresponses truly primary? Int Immunol 2:9–13. [DOI] [PubMed] [Google Scholar]
- 48.Heeger PS, Greenspan NS, Kuhlenschmidt S, Dejelo C, Hricik DE, Schulak JA, and Tary-Lehmann M 1999. Pretransplant frequency of donor-specific, IFN-gamma-producing lymphocytes is a manifestation of immunologic memory and correlates with the risk of posttransplant rejection episodes. J Immunol 163:2267–2275. [PubMed] [Google Scholar]
- 49.O’Brien RM, and Kemeny DM 1998. Distribution of alloreactivity amongst the CD45 isoforms of circulating CD4 and CD8 T lymphocytes. Scand J Immunol 48:679–683. [DOI] [PubMed] [Google Scholar]
- 50.Epperson DE, and Pober JS 1994. Antigen-presenting function of human endothelial cells. Direct activation of resting CD8 T cells. J Immunol 153:5402–5412. [PubMed] [Google Scholar]
- 51.Taylor DK, Neujahr D, and Turka LA 2004. Heterologous immunity and homeostatic proliferation as barriers to tolerance. Curr Opin Immunol 16:558–564. [DOI] [PubMed] [Google Scholar]
- 52.Adams AB, Pearson TC, and Larsen CP 2003. Heterologous immunity: an overlooked barrier to tolerance. Immunol Rev 196:147–160. [DOI] [PubMed] [Google Scholar]
- 53.Pantenburg B, Heinzel F, Das L, Heeger PS, and Valujskikh A 2002. T Cells Primed by Leishmania major Infection Cross-React with Alloantigens and Alter the Course of Allograft Rejection. J Immunol 169:3686–3693. [DOI] [PubMed] [Google Scholar]
- 54.Brehm MA, Markees TG, Daniels KA, Greiner DL, Rossini AA, and Welsh RM 2003. Direct visualization of cross-reactive effector and memory allo-specific CD8 T cells generated in response to viral infections. J Immunol 170:4077–4086. [DOI] [PubMed] [Google Scholar]
- 55.Adams AB, Williams MA, Jones TR, Shirasugi N, Durham MM, Kaech SM, Wherry EJ, Onami T, Lanier JG, Kokko KE, Pearson TC, Ahmed R, and Larsen CP 2003. Heterologous immunity provides a potent barrier to transplantation tolerance. J Clin Invest 111:1887–1895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Williams MA, Onami TM, Adams AB, Durham MM, Pearson TC, Ahmed R, and Larsen CP 2002. Cutting edge: persistent viral infection prevents tolerance induction and escapes immune control following CD28/CD40 blockade-based regimen. J Immunol 169:5387–5391. [DOI] [PubMed] [Google Scholar]
- 57.Burrows SR, Khanna R, Burrows JM, and Moss DJ 1994. An alloresponse in humans is dominated by cytotoxic T lymphocytes (CTL) cross-reactive with a single Epstein-Barr virus CTL epitope: implications for graft-versus-host disease. J Exp Med 179:1155–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Burrows SR, Khanna R, Silins SL, and Moss DJ 1999. The influence of antiviral T-cell responses on the alloreactive repertoire. Immunol Today 20:203–207. [DOI] [PubMed] [Google Scholar]
- 59.Bingaman AW, and Farber DL 2004. Memory T cells in transplantation: generation, function, and potential role in rejection. Am J Transplant 4:846–852. [DOI] [PubMed] [Google Scholar]
- 60.Nahill SR, and Welsh RM 1993. High frequency of cross-reactive cytotoxic T lymphocytes elicited during the virus-induced polyclonal cytotoxic T lymphocyte response. J Exp Med 177:317–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Welsh RM, and Selin LK 2002. No one is naive: the significance of heterologous T-cell immunity. Nat Rev Immunol 2:417–426. [DOI] [PubMed] [Google Scholar]
- 62.Ashwell JD, Chen C, and Schwartz RH 1986. High frequency and nonrandom distribution of alloreactivity in T cell clones selected for recognition of foreign antigen in association with self class II molecules. J Immunol 136:389–395. [PubMed] [Google Scholar]
- 63.Selin LK, Nahill SR, and Welsh RM 1994. Cross-reactivities in memory cytotoxic T lymphocyte recognition of heterologous viruses. J Exp Med 179:1933–1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Selin LK, Varga SM, Wong IC, and Welsh RM 1998. Protective heterologous antiviral immunity and enhanced immunopathogenesis mediated by memory T cell populations. J Exp Med 188:1705–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Welsh RM, McNally JM, Brehm MA, and Selin LK 2000. Consequences of cross-reactive and bystander CTL responses during viral infections. Virology 270:4–8. [DOI] [PubMed] [Google Scholar]
- 66.Chen HD, Fraire AE, Joris I, Brehm MA, Welsh RM, and Selin LK 2001. Memory CD8+ T cells in heterologous antiviral immunity and immunopathology in the lung. Nat Immunol 2:1067–1076. [DOI] [PubMed] [Google Scholar]
- 67.Kim SK, Brehm MA, Welsh RM, and Selin LK 2002. Dynamics of memory T cell proliferation under conditions of heterologous immunity and bystander stimulation. J Immunol 169:90–98. [DOI] [PubMed] [Google Scholar]
- 68.Selin LK, Cornberg M, Brehm MA, Kim SK, Calcagno C, Ghersi D, Puzone R, Celada F, and Welsh RM 2004. CD8 memory T cells: cross-reactivity and heterologous immunity. Semin Immunol 16:335–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Welsh RM, Selin LK, and Szomolanyi-Tsuda E 2004. Immunological memory to viral infections. Annu Rev Immunol 22:711–743. [DOI] [PubMed] [Google Scholar]
- 70.Clute SC, Watkin LB, Cornberg M, Naumov YN, Sullivan JL, Luzuriaga K, Welsh RM, and Selin LK 2005. Cross-reactive influenza virus-specific CD8+ T cells contribute to lymphoproliferation in Epstein-Barr virus-associated infectious mononucleosis. J Clin Invest 115:3602–3612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kim SK, Cornberg M, Wang XZ, Chen HD, Selin LK, and Welsh RM 2005. Private specificities of CD8 T cell responses control patterns of heterologous immunity. J Exp Med 201:523–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Cornberg M, Chen AT, Wilkinson LA, Brehm MA, Kim SK, Calcagno C, Ghersi D, Puzone R, Celada F, Welsh RM, and Selin LK 2006. Narrowed TCR repertoire and viral escape as a consequence of heterologous immunity. J Clin Invest 116:1443–1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Selin LK, Brehm MA, Naumov YN, Cornberg M, Kim SK, Clute SC, and Welsh RM 2006. Memory of mice and men: CD8+ T-cell cross-reactivity and heterologous immunity. Immunol Rev 211:164–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wu Z, Bensinger SJ, Zhang J, Chen C, Yuan X, Huang X, Markmann JF, Kassaee A, Rosengard BR, Hancock WW, Sayegh MH, and Turka LA 2004. Homeostatic proliferation is a barrier to transplantation tolerance. Nat Med 10:87–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Badovinac VP, Haring JS, and Harty JT 2007. Initial T cell receptor transgenic cell precursor frequency dictates critical aspects of the CD8(+) T cell response to infection. Immunity 26:827–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Joshi NS, Cui W, Chandele A, Lee HK, Urso DR, Hagman J, Gapin L, and Kaech SM 2007. Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 27:281–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Korngold R, and Sprent J 1990. T-Cell Subsets and Graft-vs.-Host Disease. In Graft-vs.-Host Disease. Burakoff SJ, Deeg HJ, Ferrara J, and Atkinson K, eds. Marcel Dekker, Inc., New York. 31–50. [Google Scholar]
- 78.Fecteau S, Basadonna GP, Freitas A, Ariyan C, Sayegh MH, and Rothstein DM 2001. CTLA-4 up-regulation plays a role in tolerance mediated by CD45. Nat Immunol 2:58–63. [DOI] [PubMed] [Google Scholar]
- 79.Baron V, Bouneaud C, Cumano A, Lim A, Arstila TP, Kourilsky P, Ferradini L, and Pannetier C 2003. The repertoires of circulating human CD8(+) central and effector memory T cell subsets are largely distinct. Immunity 18:193–204. [DOI] [PubMed] [Google Scholar]
- 80.Anderson BE, Taylor PA, McNiff JM, Jain D, Demetris AJ, Panoskaltsis-Mortari A, Ager A, Blazar BR, Shlomchik WD, and Shlomchik MJ 2008. Effects of donor T cell trafficking and priming site on GVHD induction by naive and memory phenotype CD4 T cells. Blood:blood-2007–2009-107953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Beilhack A, Schulz S, Baker J, Beilhack GF, Nishimura R, Baker EM, Landan G, Herman EI, Butcher EC, Contag CH, and Negrin RS 2007. Prevention of acute graft-versus-host disease by blocking T-cell entry to secondary lymphoid organs. Blood. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mazo IB, Honczarenko M, Leung H, Cavanagh LL, Bonasio R, Weninger W, Engelke K, Xia L, McEver RP, Koni PA, Silberstein LE, and von Andrian UH 2005. Bone marrow is a major reservoir and site of recruitment for central memory CD8+ T cells. Immunity 22:259–270. [DOI] [PubMed] [Google Scholar]
- 83.van Lier RA, ten Berge IJ, and Gamadia LE 2003. Human CD8(+) T-cell differentiation in response to viruses. Nature Reviews. Immunology. 3:931–939. [DOI] [PubMed] [Google Scholar]
- 84.Gamadia LE, van Leeuwen EM, Remmerswaal EB, Yong SL, Surachno S, Wertheim-van Dillen PM, Ten Berge IJ, and Van Lier RA 2004. The size and phenotype of virus-specific T cell populations is determined by repetitive antigenic stimulation and environmental cytokines. J Immunol 172:6107–6114. [DOI] [PubMed] [Google Scholar]
- 85.Appay V, Dunbar PR, Callan M, Klenerman P, Gillespie GM, Papagno L, Ogg GS, King A, Lechner F, Spina CA, Little S, Havlir DV, Richman DD, Gruener N, Pape G, Waters A, Easterbrook P, Salio M, Cerundolo V, McMichael AJ, and Rowland-Jones SL 2002. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat Med 8:379–385. [DOI] [PubMed] [Google Scholar]
- 86.Arstila TP, Casrouge A, Baron V, Even J, Kanellopoulos J, and Kourilsky P 1999. A direct estimate of the human alphabeta T cell receptor diversity. Science 286:958–961. [DOI] [PubMed] [Google Scholar]
- 87.Moon JJ, Chu HH, Pepper M, McSorley SJ, Jameson SC, Kedl RM, and Jenkins MK 2007. Naive CD4(+) T cell frequency varies for different epitopes and predicts repertoire diversity and response magnitude. Immunity 27:203–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Choi EY, Christianson GJ, Yoshimura Y, Sproule TJ, Jung N, Joyce S, and Roopenian DC 2002. Immunodominance of H60 is caused by an abnormally high precursor T cell pool directed against its unique minor histocompatibility antigen peptide. Immunity 17:593–603. [DOI] [PubMed] [Google Scholar]
- 89.Friedman TM, Goldgirsh K, Berger SA, Zilberberg J, Filicko-O’Hara J, Flomenberg N, Donato M, Rowley SD, and Korngold R 2008. Overlap between in vitro donor anti-host and in vivo post-transplantation TCR V{beta} utilization: A new paradigm for designer allogeneic blood and marrow transplantation. Blood. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Zilberberg J, McElhaugh D, Gichuru LN, Korngold R, and Friedman TM 2008. Inter-strain tissue-infiltrating T cell responses to minor histocompatibility antigens involved in graft-versus-host disease as determined by Vbeta spectratype analysis. J Immunol 180:5352–5359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.DiRienzo CG, Murphy GF, Friedman TM, and Korngold R 2007. T-cell receptor V(alpha) usage by effector CD4+Vbeta11+ T cells mediating graft-versus-host disease directed to minor histocompatibility antigens. Biol Blood Marrow Transplant 13:265–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Wettstein PJ, and Bailey DW 1982. Immunodominance in the immune response to “multiple” histocompatibility antigens. Immunogenetics 16:47–58. [DOI] [PubMed] [Google Scholar]
- 93.Korngold R, and Wettstein PJ 1990. Immunodominance in the graft-vs-host disease T cell response to minor histocompatibility antigens. J Immunol 145:4079–4088. [PubMed] [Google Scholar]
- 94.Choi EY, Christianson GJ, Yoshimura Y, Jung N, Sproule TJ, Malarkannan S, Joyce S, and Roopenian DC 2002. Real-time T-cell profiling identifies H60 as a major minor histocompatibility antigen in murine graft-versus-host disease. Blood 100:4259–4265. [DOI] [PubMed] [Google Scholar]
- 95.Krenger W, and Ferrara JL 1996. Graft-versus-host disease and the Th1/Th2 paradigm. Immunol Res 15:50–73. [DOI] [PubMed] [Google Scholar]
- 96.van den Brink MR, and Burakoff SJ 2002. Cytolytic pathways in haematopoietic stem-cell transplantation. Nat Rev Immunol 2:273–281. [DOI] [PubMed] [Google Scholar]
- 97.Shlomchik WD 2007. Graft-versus-host disease. Nat Rev Immunol 7:340–352. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.







