Alloantigen Expression on Host Target Epithelium Inhibits Graft-Versus-Leukemia (GVL) Effects.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 596-596
Author(s):  
Shoji Asakura ◽  
Daigo Hashimoto ◽  
Ken-ichi Matsuoka ◽  
Yukimi Sakoda ◽  
Mitsune Tanimoto ◽  
...  

Abstract We previously demonstrated that alloantigen expression on host target epithelium is not necessary but augment acute GVHD (Nat Med 2002). Here, we tested the role of alloantigen expression on host target epithelium on the GVL effect. We created BM chimeras, [B6 → C3H.Sw] where only host hematopoietic cells express multiple minor histocompatibility antigens (mHAs) allogeneic to the donors but not on host target epithelium. Identically treated [B6 → B6] chimeras were created as controls where both hematopoietic and target cells express mHAs allogeneic to the donors. Four month later, these chimeras were reirradiated and were injected with 5×106 BM and 1×106 CD8 T cells harvested from C3H.Sw donors. Acute GVHD developed in [B6 → C3H.Sw] chimeras but was less severe in these chimeras than controls (Table). Next, animals were transplanted, as above, with the addition of 2500 B6-derived EL4 thymoma to the donor inoculum. The cause of death was determined by postmortem examination to be either GVHD or leukemia (presence of hepatic and/or splenic nodules). All [B6 → B6] recipients of C3H.Sw CD8+ cells died from leukemia, although their survival time was significantly prolonged compared to syngeneic controls (P<.01). Surprisingly, [B6 → C3H.Sw] chimeras displayed more potent GVL effects than controls (Table), in spite of reduced GVHD in these chimeras. Similar results were obtained in the other sets of chimeras [DBA → Balb/c] when injected with BM and T cells isolated from Balb/c donors together with DBA-derived P815 mastocytoma and [Balb/c → DBA] when injected with BM and T cells isolated from DBA donors and Balb/c-derived A20 lymphoma. These animals displayed more potent GVL effects compared to control chimeras (Table). To elucidate the mechanisms of this superior GVL effect in these chimeras, analysis of the spleen was performed 3 weeks after BMT. Expansion and activation of donor CD8+ T cells were greater in [B6 → C3H.Sw] recipients than those in [B6 → B6] recipients in spleens (P<.05). Thus, alloantigens expressed on host antigen-presenting cells stimulate host-reactive T cells but in the absence of alloantigen expression on host epithelium contraction of host-reactive T cells may be impaired, resulting in a superior GVL effect. These results provides a complete picture of the role of alloantigen expression on host epithelium in allogeneic hematopoietic cell transplantation; alloantigen expression on host target cells i) does not always necessary to induce acute GVHD, ii) augment GVHD, and iii) suppress GVL effects. These imply that allogeneic cellular therapy targeting at mHA preferentially expressed on hematopoietic cells may induce potent GVL effects while inducing less severe GVHD. Donor Recipients GVHD score (d21) GVHD mortality (d50) Leukemia mortality (d50) *P<.05. **not examined B6 CD8 [B6 → B6] 1.0 +/− 0.4 0% 100% C3H.Sw CD8 [B6 → B6] 3.7 +/− 0.3 33% 95% C3H.Sw TCD [B6 → C3H.Sw] NE** 0% 100% C3H.Sw CD8 [B6 → C3H.Sw] 2.1 +/− 0.4* 0%* 69%* Balb/c TCD [DBA → DBA] 0.2 +/− 0.3 NE** 100% Balb/c T [DBA→ DBA] 3.2 +/− 0.6 NE** 60% Balb/c TCD [DBA→ Balb/c] NE** NE** 100% Balb/c T [DBA→ Balb/c] 1.3 +/− 0.3* NE** 10%* DBA TCD [Balb/c→ Balb/c] 0.0 +/− 0.0 NE** 100% DBA T [Balb/c→ DBA] 1.8 +/− 0.6 NE** 100% DBA TCD [Balb/c→ Balb/c] NE** NE** 100% DBA T [Balb/c→ DBA] 0.8 +/− 0.3 NE** 30%*

2011 ◽  
Vol 208 (5) ◽  
pp. 1069-1082 ◽  
Author(s):  
Daigo Hashimoto ◽  
Andrew Chow ◽  
Melanie Greter ◽  
Yvonne Saenger ◽  
Wing-Hong Kwan ◽  
...  

Acute graft-versus-host disease (GVHD) results from the attack of host tissues by donor allogeneic T cells and is the most serious limitation of allogeneic hematopoietic cell transplantation (allo-HCT). Host antigen-presenting cells are thought to control the priming of alloreactive T cells and the induction of acute GVHD after allo-HCT. However, whereas the role of host DC in GVHD has been established, the contribution of host macrophages to GVHD has not been clearly addressed. We show that, in contrast to DC, reducing of the host macrophage pool in recipient mice increased donor T cell expansion and aggravated GVHD mortality after allo-HCT. We also show that host macrophages that persist after allo-HCT engulf donor allogeneic T cells and inhibit their proliferation. Conversely, administration of the cytokine CSF-1 before transplant expanded the host macrophage pool, reduced donor T cell expansion, and improved GVHD morbidity and mortality after allo-HCT. This study establishes the unexpected key role of host macrophages in inhibiting GVHD and identifies CSF-1 as a potential prophylactic therapy to limit acute GVHD after allo-HCT in the clinic.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2015-2015
Author(s):  
Vincent Schwarze ◽  
Anne-Kathrin Hechinger ◽  
Franziska Leonhardt ◽  
Gabriele Prinz ◽  
Annette Schmitt-Gräff ◽  
...  

Abstract Allogeneic hematopoietic cell transplantation (allo-HCT) is an important treatment option for different hematological malignancies, but also for some nonmalignant hematological disorders such as sickle cell anemia, aplastic anemia or thalassemia(1). In the ladder group the graft-versus-leukemia (GvL) effect mediated by donor T cells is less important and prevention of graft-versus-host disease (GvHD), which occurs in 40-50% of allo-HCT patients, is a major priority. The common gamma chain (CD132) is a cytokine receptor sub-unit that is common to the interleukin (IL) receptors of IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21. Since several of these cytokines were shown to be increased in the serum of patients developing acute GvHD, we reasoned that inhibition of CD132 could have a profound effect on acute GvHD by inhibiting the bioactivity of multiple proinflammatory cytokines. We observed that anti-CD132 treatment reduced GvHD potently with respect to survival, production of TNF, IFN-γ, IL-6, MCP-1 and GvHD histopathology. Protection was only seen when anti-CD132 was applied in a CD8 T cell-dependent GvHD model while no protection was seen when only CD4 T cells were given. Mechanistically, we could show that CD8 T cells isolated from mice treated with anti-CD132 had reduced levels of Granzyme B and that GvHD induced by Perforin-deficient T cells was resistant towards blockade by anti-CD132 treatment. These data indicated a role of the common gamma chain cytokines for the induction of Perforin/Granzyme B in CD8 T cells during GvHD. Compatible with this notion, exposure of CD8 T cells towards IL-2, IL-7, IL-15 and IL-21 alone or in combination induced increased levels of Granzyme B. Based on these findings, we concluded that CD8 T cells that are activated by common gamma chain cytokines during GvHD produce then Granzyme B which can be blocked by anti-CD132 treatment. This therapeutic approach has particular clinical potential for patients undergoing allogeneic transplantation for nonmalignant indications, since graft-versus-tumor activity is not required. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (10) ◽  
pp. 3393-3399 ◽  
Author(s):  
Chang-Ki Min ◽  
Yoshinobu Maeda ◽  
Kathleen Lowler ◽  
Chen Liu ◽  
Shawn Clouthier ◽  
...  

Abstract Administration of exogenous interleukin-18 (IL-18) regulates experimental acute graft-versus-host disease (GVHD) in a Fas-dependent manner when donor CD4+ T cells are required for mortality after experimental allogeneic bone marrow transplantation (BMT). However, CD4+ and CD8+ T cells can induce acute GVHD after clinical allogeneic BMT, and the role of IL-18 in CD8+-mediated acute GVHD is unknown. We, therefore, determined the role of IL-18 in GVHD mediated by CD4+ or CD8+ T cells across major histocompatibility complex (MHC) class II- and class I-disparate allogeneic BMT, respectively. Administering IL-18 significantly increased survival in CD4+-mediated GVHD but reduced survival in CD8+-mediated GVHD. This increase in deaths was associated with significantly greater clinical, biochemical, and histopathologic parameters of GVHD damage and was independent of Fas expression on donor T cells. Administering IL-18 significantly enhanced allospecific cytotoxic function and expansion of CD8+ cells. Endogenous IL-18 was critical to GVHD mediated by CD8+ donor T cells because IL-18 receptor-deficient donors caused significantly less GVHD but exacerbated CD4+-mediated, GVHD-related death. Furthermore, administering anti-IL-18 monoclonal antibody significantly reduced CD8+-mediated, GVHD-related death. Together these findings demonstrate that IL-18 has paradoxical effects on CD4+ and CD8+ cell-mediated GVHD. (Blood. 2004;104:3393-3399)


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1260-1260
Author(s):  
Tania Kollgaard ◽  
Soren L. Petersen ◽  
Sine Reker Hadrup ◽  
Tania N. Masmas ◽  
Tina Seremet ◽  
...  

Abstract Allogeneic hematopoietic cell transplantation (HCT) has a well-documented ability to cure a number of malignant hematological diseases. The curative principle in allogeneic HCT is the Graft-versus-Leukemia (GVL) effect and nonmyeloablative (NMA) conditioning HCT relies exclusively on this anti-tumor effect to eliminate tumor cells. Donor T-cells are documented to be responsible for the GVL effect, however, often they also cause Graft-versus-Host disease (GVHD) which is associated with high morbidity and mortality. Characterization of cells and molecules involved in both GVL and GVHD would potentially set the stage for separation of GVL and GVHD in order to augment GVL in the absence of GVHD. In the present study, we analyzed the clonotype composition of CD8+ T cells following NMA conditioning and HCT, in two patients with chronic lymphocytic leukemia (CLL). T-cell receptor (TCR) clonotype mapping (RT-PCR combined with denaturing gradient gel electrophoresis (DGGE)) was used to identify clonally expanded CD8+ T cells in blood samples. This method provides a “molecular fingerprint” of each unique T cell based on junctional diversity of the TCR CDR3 region and, thus, offers the means to track T-cell clonotypes in time and space. Longitudinal comparative analyses showed that CD8+ T-cell clonality was highly dynamic during early phases after transplantation with various clonotypes emerging and disappearing. However, clonal diversity decreased after 4–5 months and stable CD8+ T-cell clonotypes appeared and persisted throughout the analyzed period (up to two years). One patient received donor lymphocyte infusion (DLI) due to disease progression and this was shown to lead to establishment of recurrent (detected prior to DLI) CD8+ T-cell clonotypes as well as new CD8+ T-cell clonotypes. The appearance of these cells correlated with disease remission strongly suggesting their engagement in anti-CLL reactivity. To examine the functional capacities of clonally expanded T cells after HCT, recipient CD8+ T cells were stimulated ex vivo with pre-transplant patient CLL cells and/or normal hematopoietic cells and the T-cell surface expression of CD107a (marker for cytotoxicity) was detected by FACS. Clonotype mapping analyses of FACS sorted CD107a positive CD8+ T cells after stimulation with CLL cells demonstrated that such cytotoxic CD8+ T cells were present as stable clonally expanded T cells in vivo strongly implying their involvement in an ongoing anti-CLL-response. Furthermore, co-culture with normal hematopoietic cells resulted in a unique CD107a positive expanded T-cell clonotype. Our results strongly suggest that clonally expanded CD8+ T cells are involved in an ongoing tumor response and support data which demonstrate that GVL and GVHD are the result of distinct responses.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2008-2008
Author(s):  
Mohammad Sohrab Hossain ◽  
Vicky F El-Najjar ◽  
David L Jaye ◽  
Rafi Ahmed ◽  
Edmund K. Waller

Abstract Background Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative treatment for patients with both malignant and non-malignant hematologic disorders. But life-threatening graft-vs-host diseases (GvHD) caused by alloreactive donor T cells limits its clinical use. Alloreactive T cells are also required for graft-vs-leukemia (GvL) and to fight opportunistic infections. Hence, a method that modulates donor T cells activity to reduce GvHD but to retain GvL effect is highly desirable. The inhibitory receptor programed cell death-1 (PD-1) reduces T cell activation through binding with its ligand PD-L1 or PD-L2. Interaction between PD-1 and PD-L1 induces cardiac allograft tolerance and expression of PD-L1 is upregulated in presence of inflammatory stimuli. Here, we studied the role of PD-L1 expression on hematologic and non-hematologic tissues and PD-1 - PD-L1 binding in the development of GvHD. Methods Wild type C57BL/6 (WT B6), PD-L1 knock out B6 (KO) and PD-L2 KO B6 mice were transplanted with 2 x106 splenic T cells and bone marrow (BM) cells from H-2K B10.BR donors. The average acute GvHD scores were determined by combining the GvHD scores obtained from the histological tissue sections of small intestine, large intestine and liver, and weight-loss, posture, activity, fur texture and skin integrity data following standard published procedures. The activation status of splenic T cells was analyzed by flow cytometry. Serum cytokines were determined by using 26 plex Luminex assay. The requirement of hematopoietic and or non-hematopoietic tissues expressing PD-L1 to reduce GvHD was investigated by generating radiation chimeras using WT B6 mice engrafted with PD-L1 KO BM and vice versa. Two months later radiation chimeras were transplanted again with 2 x106 splenic T cells along with 2 x106 BM cells from congenic na•ve H-2K donors. The role of PD-L1 expressing donor hematopoietic cells on the development of acute GvHD was tested by transplanting B10.BR mice with donor PD-L1 KO B6 and WT B6 splenocytes. Results PD-L1 KO B6 recipients had significantly increased acute GvHD (scores 1.68 ± 0.07) compared with WT B6 GvHD (0.78 ± 0.024, p<0.0005) and B6 PD-L2 KO B6 (0.86 ± 0.14, p<0.0005) within day 8 after transplant. All PD-L1 KO B6 recipients had severe GvHD with >25% weight loss on day 8 after transplant and were sacrificed. The WT B6 and PD-L2 KO B6 recipients survived 75% and 80%, respectively until 34 days of transplantation with similar levels of chronic GvHD. To test whether excessive activation of donor T cells caused severe acute GvHD in PD-L1 KO B6 recipients, we determined the activation status of donor T cells in the spleen. The numbers of donor CD4+ and CD8+ T cells expressing ICOS-1 and PD-1 in the spleen were significantly higher (p<0.005) in PD-L1 KO B6 recipients compared with the WT B6 and PD-L2 KO B6 recipients. Additionally, significantly increased levels of serum inflammatory cytokines (IFN-g and TNF-a) were also detected in the PD-L1 KO B6 recipients compared with the WT B6 recipients on day 8 post transplant (Figure 1). Using WT B6 or PD-L1KO hematopoietic cell radiation chimeras as allo-HSCT recipients, we further confirmed that both allo-HSCT radiation chimeras having PD-L1 expressing hematopoietic (10% survival, open square) and non-hematopoietic cells (10% survival, closed triangle) were required to protect from GvHD (Figure 2). We next investigate whether PD-L1 KO donor cells cause increased GvHD. The B10.BR recipients transplanted with donor PD-L1 KO B6 splenocytes had 70% survival while the same recipients transplanted with WT B6 donor splenocytes had only 20% survival until 100 days post transplant. These data suggest that only PD-L1 expressed by host tissues is required to inhibit the development of GvHD. In summary, our data suggest that PD-L1 expressed by host tissues are required to control GvHD and method(s) that enhance expression of PD-L1 in allo-HSCT recipients may represent a novel strategy to control GvHD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2146-2146
Author(s):  
Shoubao Ma ◽  
Huanle Gong ◽  
Shuangzhu Liu ◽  
Jingjing Han ◽  
Zhinan Yin ◽  
...  

Abstract IL-27 is a member of the IL-12 cytokine family that consists of EBV-induced gene 3 (EBI3), and p28. IL-27 is produced by activated antigen-presenting cells such as dendritic cells (DCs) and macrophages, and it signals through a heterodimeric receptor (IL-27R) consisting of the WSX-1 and the gp130. Emerging evidence has shown that IL-27 has both pro- and anti-Inflammatory effects. Recent study demonstrated a protected role IL-27p28 in acute graft-versus-host disease (aGVHD) in parent-to-F1 murine aGVHD model (Marillier et al., Eur. J. Immunol. 2014). However, the precise role of IL-27 in the development of aGVHD remains largely unknown. In this study, we carried out studies in a murine aGVHD model of fully MHC-mismatched myeloablative bone marrow transplantation (C57BL/6 to BALB/c). Lethally irradiated BALB/c mice transplanted with WT B6 bone marrow (BM) plus IL-27-/-(IL-27p28flox/flox-CD11c-cre (Itgax-p28f/f)) splenocytes had significantly accelerated aGVHD mortality comparing with those received WT BM plus splenocytes (P<0.001). However, similar aGVHD mortality was observed between mice transplanted with WT BM plus WT splenocytes and IL-27-/-BM plus WT splenocytes, and IL-27-/-BM plus IL-27-/- splenocytes, suggesting that IL-27 from splenocytes, not BM-derived cells, had protective effect in aGVHD. Additionally, mice were transplanted with IL-27-/- BM and splenocytes, and then treated with rmIL-27 on days 0 and 7 post BMT. rmIL-27 treated mice had longer survival and alleviated the clinical signs of aGVHD compared to PBS-treated mice (P<0.05). We next explored the mechanisms by which IL-27 protects mice against aGVHD. We found the expression of the cell surface IL-27 receptor, WSX-1 and gp130, were increased on both CD4+ and CD8+T cells after allo-stimulation. Moreover, IL-27 inhibited cell proliferation of allo-reactive T cells in mix lymphocytes reactivation (MLR) in vitro. We also found that the percentages of CD69+CD4+, CD69+CD8+T cells, as well as IFN-γ expression by CD4+ and CD8+T, were significantly increased in the spleen, liver and intestinal intraepithelial lymphocytes (IEL) from mice transplanted with WT BM and IL-27-/- splenocytes. Furthermore, we observed increased frequencies and numbers of Treg cells and MDSCs in aGVHD target organs in mice reconstituted with IL-27-/-splenocytes. Together, these results indicated that IL-27 alleviated aGVHD may through suppressing Th1 cell responses and promoting immune suppressing cells, including MDSCs and Treg cells. To further clarify the role of IL-27 in aGVHD, mice were transplanted with WT BM plus IL-27R-/-(B6N.129P2-Il27ratm1Mak/J) splenocytes, the results, however, showed that IL-27R deficiency in donor T cells significantly attenuated aGVHD, which was unpredicted, and was contrary to that of IL-27 deficiency results. Additional studies showed that IL-27R deficiency in T cells inhibited allo-reactive T cells proliferation and IL-2, IFN-γ production in MLR assay. The percentages of CD69+ T cells and IFN-γ+ CD4+ and CD8+T were significantly decreased, while the MDSCs and Treg cells wereincreased in aGVHD target organs from mice transplanted IL-27R-/-splenocytes. These results indicated that lack of IL-27R signaling resulted in downregulation of intrinsic immune responses which led to alleviation of aGVHD. Previous study reported that soluble form of IL-27Ra is highly existed in human serum and may severs as a natural IL-27 antagonist (Dietrich et al., JI. 2014). We hypothesized that sIL-27Ra may function as a decoy receptor to regulate aGVHD through inhibiting IL-27 signaling. To test this hypothesis, WT recipients were given exogenous soluble mouse IL-27 Ra Fc chimera protein known to block the binding of IL-27 to the membrane bound form of IL-27R. WT recipients given IL-27 Ra-Fc to block IL-27R/IL-27 interaction had significantly exacerbated GVHD mortality. The percentages of CD69+ T cells and IFN-γ+ CD4+ and CD8+T were significantly increased in aGVHD target organs from mice received IL-27 Ra fusion protein. In conclusion, this is the first demonstration in the same allogeneic BMT model that IL-27 deficiency augments but IL-27R deficiency alleviates acute GVHD. Our studies provide further evidence for the protective role of rmIL-27, and promoting effect of sIL-27R in the same aGVHD model. This study warrants further investigations of the possible therapeutic applications of IL-27 in acute GVHD. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 62-62 ◽  
Author(s):  
Ying Chen ◽  
Chia-Lei Lin ◽  
Nainong Li ◽  
Gong Du ◽  
Lieping Chen ◽  
...  

Abstract We reported that donor CD8+ T cells mediated graft versus leukemia (GVL) activity without causing graft versus host disease (GVHD) in anti-CD3-conditioned recipients, although the same dose of donor CD8+ T cells caused lethal GVHD in recipients conditioned with sublethal total body irradiation (TBI) (J. Immunol. 2007). We recently observed that donor CD8+ T cells from the liver of anti-CD3-conditioned recipients showed a marked reduction in proliferation in response to anti-CD3/CD28 stimulation, as compared to those from TBI-conditioned recipients, indicating that donor CD8+ T cells are tolerized in the tissues of anti-CD3-conditioned recipients. B7H1, a co-inhibitory molecule, is constitutively expressed by hematopoietic cells and expressed by parenchymal cells after IFN-γ induction. B7H1 tolerizes T cells by interaction with its ligand PD-1 on activated T cells. To explore the role of B7H1 in GVHD prevention in anti-CD3-conditioned recipients, donor C57BL/6 CD8+ T cells (20×106) and bone marrow cells (100×106) were transplanted into anti-CD3-conditioned wild-type or B7H1−/− BALB/c recipients. While the wild-type recipients all survived for more than 100 days without signs of GVHD, the B7H1−/− recipients developed severe GVHD with diarrhea, weight-loss, and hair-loss, and 70% of them died 60 days after transplantation. Similarly, while donor CD8+ T and BM cells induced little GVHD in unconditioned Rag-2−/− BALB/c recipients, they induced severe lethal GVHD in B7H1−/− Rag-2−/− BALB/c recipients. Furthermore, donor CD8+ T and BM cells still induced lethal GVHD in unconditioned chimeric B7H1−/−Rag-2−/− recipients reconstituted with B7H1+/+Rag-2−/− BM. In addition, we observed upregulation of B7H1 expression by hepatocytes and intestine epithelial cells of anti-CD3-conditioned BALB/c or unconditioned Rag-2−/− recipients after donor cells infusion, as judged by RT-PCR, flow cytometry analysis, and histoimmunostaining of B7H1. In vivo bioluminescent imagine showed much more severe tissue infiltration of donor T cells in B7H1−/− recipients as compared to B7H1+/+ recipients, and the in vitro proliferation of donor CD8+ T cells from the liver of the former recipients was much more vigorous than that of the latter recipients. These results demonstrate that B7H1 expression by tissue parenchymal cells rather than hematopoietic cells tolerizes infiltrating donor T cells in GVHD target tissues and prevents GVHD; and that the radiation-free anti-CD3-conditioning regimen can maintain this tissue protection mechanism. (This study is surpported by Marcus Foundation and NIH R01 AI 066008).


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2975-2975
Author(s):  
Isao Tawara ◽  
Yaping Sun ◽  
Chen Liu ◽  
Tomomi Toubai ◽  
Pavan Reddy

Abstract Abstract 2975 IL-10 is a key immune-regulatory cytokine and its gene polymorphisms correlate with severity of clinical graft-versus-host (GVHD). IL-10 is made by a variety of donor and host cells, but the functional relevance of its source in acute GVHD is not well understood. We utilized well characterized MHC matched, minor mismatched C3H.SW →B6 model of acute GVHD to first examine the relevance host hematopoietic derived IL-10 on acute GVHD induced by alloreactive donor T effector cells in the absence of donor regulatory T (Tregs) cells. We generated [WTB6→B6-Ly5.2] and [IL-10−/−B6→B6-Ly5.2] chimeras such that only the host hematopoietic derived cells were incapable of producing IL-10 and utilized them as recipients. The chimeras were lethally irradiated and transplanted with 5×106 T cell-depleted (TCD) BM and 2×105 CD8+ T cells from either the allogeneic C3H.SWor the syngeneic B6 donors. All of the allogeneic animals demonstrated similar clinical severity and mortality from GVHD (65% vs 72%, P=NS). Because IL-10 can also be derived from non-hematopoietic cells, we next determined whether the absence of IL-10 secretion by both hematopoietic and non-hematopoietic cells would modulate GVHD severity. To this end, the WT B6 and the IL-10−/− B6 animals were irradiated with 10Gy and transplanted 5×106 T cell-depleted bone marrow (TCDBM) and 2×105 CD8+ T cells from either allogeneic C3H.SW or the syngeneic B6 donors. All of the allogeneic animals demonstrated similar severity and mortality from GVHD regardless of whether all of the host cells can make IL-10 (P=0.1, NS). We next examined relevance of IL-10 deficiency in donor cells. 1×106 CD25+depleted donor T effectors from either WT or IL-10−/− B6 mice were infused along with 5×106 TCDBM from WT B6 into lethally irradiated allogeneic C3H.SW or syngeneic B6 animals. The allogeneic animals that received either WT or IL-10−/− B6 effector cells demonstrated similar GVHD severity and mortality (P=0.6). Further, when T effectors from WT B6 mice were infused along with TCDBM from either WT or IL-10−/− B6 donors, the allogeneic C3H.SW animals once again demonstrated similar GVHD mortality (70% vs 80%, P=NS). Thus deficiency of IL-10 in either host tissues or in the donor grafts did not alter CD25+ depleted donor T effector cell mediated severity of GVHD. We next examined the relevance of the source of IL-10 in donor natural (Treg) mediated regulation of GVHD. Donor CD4+CD25+ Tregs from C3H.SW donors induced equivalent protection from GVHD mortality in both WT and IL-10−/− hosts (90% vs. 100%, P=NS). To examine the relevance of IL-10 derived from Tregs, we transplanted 0.25×106 CD4+CD25+ Tregs from either B6 or IL-10−/− donors along with 1×106 CD25 depleted T effectors (1 :4 ratio) into C3H.SW animals. 100% of the allogeneic animals that received WT donor Tregs survived while only 60% of those that receive IL-10−/− donor Tregs survived (P=0.001). However, when the Il-10−/− donor Treg numbers were increased to 1×106 (1 :1 ratio), all of the allogeneic animals survived (P=0.008) demonstrating that donor Treg derived IL-10 is not obligatory but is necessary for optimal reduction of GVHD. To determine the role of donor BM derived IL-10 in Treg mediated protection from GVHD, Tregs and T effector cells from WT B6 donors (1 :4 ratio) were transplanted along with 5×106 TCD BM from either WT or IL-10−/− B6 donors into lethally irradiated allogeneic C3H.SW animals. The allogeneic recipients of WT BM were protected from GVHD mortality, donor Tregs failed to protect the allogeneic animals that received IL10−/− donor BM (85% vs. 20%, P<0.04) in whom the rate of mortality was similar to the animals that did not receive any donor Tregs (P=0.96). Mechanistic studies demonstrated that donor Tregs continued to express similar levels of foxp3 and expansion early (day 7) after BMT. However the their expansion was significantly reduced at later time-points (day 14 and 21) when the hosts received BM from IL-10−/− donors. Together these data suggest that IL-10 from either host or donors does not contribute to the induction of GVHD severity by the T effector cells. However, only donor BM and donor Treg derived IL-10 is important for donor Treg mediated suppression of GVHD. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document