The T Cell Cytolytic Molecules Fas Ligand and TRAIL, the Trafficking Molecules CCR9, β7 Integrin and PSGL-1, and the Immune Modulating Molecules OX40, CEACAM1, and CTLA4 Are Required for Thymic Graft-Versus-Host Disease

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 65-65 ◽  
Author(s):  
Il-Kang Na ◽  
Sydney X. Lu ◽  
Gabrielle L. Goldberg ◽  
Daniel Daniel Hirschhorn-Cymerman ◽  
Christopher G. King ◽  
...  

Abstract Although thymic graft-versus-host-disease (tGVHD) has been recognized as an important contributor to impaired T cell reconstitution, limited T cell repertoire and increased infection risk in patients with GVHD, the molecular basis of interactions between donor alloreactive T cells, donor bone marrow (BM)-derived thymocytes, and host hematopoietic and non-hematopoietic thymic stromal cells in GVHD has not been well-defined. Here we analyzed the role of molecules relevant for T cell trafficking, cytolytic function, and co-stimulation and co-inhibition of alloreactive T cells in tGVHD. We first demonstrated that thymic output (as measured by RAG2+ splenic recent thymic emigrants) as well as the thymic cellularity (especially of CD4+CD8+ thymocytes) were inversely proportional to numbers of mature donor T cells infused with the allograft, suggesting that tGVHD severity was inversely associated with thymic function. We then studied the migration of alloreactive donor T cells in vivo with bioluminescence imaging (BLI) and found that luciferase-expressing donor T cells infiltrated the thymus within one week after allogeneic bone marrow transplantation (BMT) (Fig. 1). Upon adoptive transfer of CFSE-labeled donor T cells we noted that thymus-infiltrating alloreactive donor T cells were largely fast-proliferating (CFSElo) and highly activated (CD25+ CD44+). We analyzed the importance of T cell trafficking molecules for tGVHD using mice deficient for certain trafficking molecules, and assessed tGVHD by loss of BM-derived CD4+CD8+ thymocytes. We found that CCR9, b7 integrin subunit, and PSGL-1 were all partially required for tGVHD, while L-selectin and aE integrin subunit may be dispensable (Fig. 2A). Similarly, we examined the role of T cell cytolytic pathways for tGVHD, and found that FasL and TRAIL were required for tGVHD, but that perforin and TNF were dispensable (Fig. 2B). Finally, we assessed the role of various T cell co-stimulatory and co-inhibitory molecules for tGVHD, and found that CEACAM1, OX40 and CTLA4 were required, while GITR was partially required and ICOS was dispensable (Fig. 2C). Upon further analysis of donor BM-derived thymocytes, we observed that Bcl-2 expression in donor BM-derived thymocytes was decreased in recipients with GVHD vs. those without GVHD, which suggests that survival of thymocytes is decreased during tGVHD. Hollander and others have previously demonstrated in non-irradiated GVH reaction models that host non-hematopoietic thymic stroma may be an important target for donor alloreactive T cells. We assessed the expression of the death receptors Fas and DR5 in thymic stroma from normal and irradiated (850 cGy) BALB/c mice. We observed that in particular, MHC class II-negative stroma (endothelial cells and fibroblasts), as well as a population of MHC class II-intermediate stroma (epithelial cells) upregulated the expression of both Fas and DR5 after irradiation. Our study defines the specific pathways for cytolysis, trafficking and immune modulation involved in tGVHD and suggests selective therapeutic targets to attenuate tGVHD and improve post-transplant T-cell reconstitution in patients with GVHD. Fig 1. BLI demonstrate a distinct distribution pattern for alloreactive donor T cells in allogeneic BMT recipients, Allogeneic Balb/c recipients show a strong signal on day 4 post-transparent after transfer of 10×108 luc+ splenocytes as measured by total body photon emission. Ex vivo imaging confirms the infiltration of luc+ splenocytes to the thymus. Fig 1. BLI demonstrate a distinct distribution pattern for alloreactive donor T cells in allogeneic BMT recipients, Allogeneic Balb/c recipients show a strong signal on day 4 post-transparent after transfer of 10×108 luc+ splenocytes as measured by total body photon emission. Ex vivo imaging confirms the infiltration of luc+ splenocytes to the thymus. Fig 2. We assessed the role of molecules relevant for T cell trafficking (A), cytolytic function (B), and co-stimulation, co-inhibition (C). Irradiated BALB/c mice received 5×106 T cell depleted C57BL/6 bone marrow + 0.25×106 purified splenic T cells. Absolute numbers of donor-BM-derived CD4+CD8+ thymocytes are shown. Black bars indicate means. p-values were calculated vs. recipients of WT T cells(*p<0.05, **p<0.01) Fig 2. We assessed the role of molecules relevant for T cell trafficking (A), cytolytic function (B), and co-stimulation, co-inhibition (C). Irradiated BALB/c mice received 5×106 T cell depleted C57BL/6 bone marrow + 0.25×106 purified splenic T cells. Absolute numbers of donor-BM-derived CD4+CD8+ thymocytes are shown. Black bars indicate means. . / p-values were calculated vs. recipients of WT T cells(*p<0.05, **p<0.01)

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4283-4283
Author(s):  
Bader Alahmari ◽  
Jaebok Choi ◽  
Matthew L Cooper ◽  
Kiran R. Vij ◽  
Julie Ritchey ◽  
...  

Abstract Acute graft versus host disease (aGvHD) is a life-threatening condition that complicates allogeneic hematopoietic cell transplantation (allo-HCT). Donor T cells recognize the recipientÕs tissues as foreign causing GvHD, however, these same donor T cells are also responsible for the beneficial graft versus leukemia (GVL). Distinguishing between GvHD and GvL is crucial for the development of safe and effective therapies following allogeneic bone marrow transplantation. We have recently reported that the disruption of alloreactive donor T cell trafficking to the GvHD target organs significantly reduces GvHD in both an MHC fully-mismatched and a minor-mismatched allo-HCT models (Choi et al., Blood 2012). In this study, we examined if inhibition of α4, which is required for transendothelial migration and access to lymphoid system and GvHD target organs, could reduce GVHD. On T cells α4 integrin subunit can form a heterodimer with either β1 to form α4β1 integrin (VLA-4) or β7 to form α4β7 (LPAM-1). To genetically eliminate α4 from allogeneic donor T cells, we generated Tie-2 cre+ α4fl/fl mice (B6, H-2b, CD45.2+). Splenic pan T cells were isolated from these mice and T cell-depleted bone marrow cells (TCD BM) isolated from congenic B6 mice (CD45.1+). 5x106 TCD BM and 5x105 splenic pan T cells were transplanted into lethally irradiated (900 cGy) allogeneic Balb/c recipient mice (H-2d, CD45.2+). We found that α4 deficient T cells significantly reduced GvHD compared to Tie-2 cre+ control T cells (Fig. A). α4 deficient T cells had no significant effect on donor engraftment and achieved complete donor chimerism. In addition, recipients transplanted with α4 deficient T cells had significantly lower histopathology score (median score 3 vs. 6; p=0.0263). Similar results were observed when donor T cells were infused at day 11 post allo-HCT. To test whether α4 deficient T cells maintain GvL we performed bioluminescence imaging (BLI) using a systemic leukemia mouse model. CBRluc-expressing A20 leukemia cells (Balb/c-derived) were transplanted intravenously (1 × 105 cells) along with TCD BM (B6, CD45.1+) into Balb/c recipients at day 0. Pan T cells (B6, CD45.2+) either from Tie-2 cre+ or α4 deficient mice were infused at day 11 to allow sufficient time for the leukemia cells to expand. After weekly BLI, we found α4 deficient T cells were able to control leukemia cells as effectively as Tie-2 cre+ Control T cells (p=0.3748). In addition, we performed BLI to track CBRluc-transduced pan T cells (2 × 106 cells) after allo-HCT in vivo. We found a significant difference in the percentage of BLI signal intensity between control and α4 deficient T cells in spleen and gut at day 14 and 21 post allo-HCT. (Fig. B) While we speculate defective T cell trafficking to GvHD target organs would be the primary reason for the reduced GvHD in the recipients of α4 deficient T cells, we examined other possible mechanisms such as T cell alloreactivity and proliferation. We found that α4 deficient T cells proliferated at the same rate as Tie-2 cre+ T cells in the presence of anti-CD3/CD28 antibody-coated beads. However, α4 deficient CD8 T cells proliferate slower in the presence of allogeneic antigen presenting cell compared to Tie-2 cre+ T cells. Interestingly, α4 deficient T cells significantly upregulate CTLA-4 and GZMB compared to Tie-2 cre+ control T cells. These data suggest that not only altered T cell trafficking to GvHD target organs but also altered T cell functions might be the reason for the reduced GvHD. In conclusion, we propose that α4 represents a promising therapeutic target for future efforts to mitigate GvHD after allo-HCT. In addition, this strategy can be exploited in other diseases besides GvHD such as solid organ transplantation, chronic inflammatory diseases and autoimmune diseases. Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 455-455
Author(s):  
Jaebok Choi ◽  
Edward Dela Ziga ◽  
Julie Ritchey ◽  
Lynne Collins ◽  
Julie Prior ◽  
...  

Abstract Abstract 455 Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative treatment for patients with relapsed/refractory leukemia, and marrow failure states such as myelodysplasia and aplastic anemia. However, allo-HSCT is complicated by allogeneic donor T cell-mediated graft-versus-host disease (GvHD) which can be life-threatening especially in recipients of unrelated or HLA-mismatched hematopoietic stem cell products. These same alloreactive donor T cells also mediate a beneficial graft-versus-leukemia (GvL) effect. Thus, the clinical goal in allo-HSCT is to minimize GvHD while maintaining GvL. Recent studies have suggested that this might be achieved by infusing regulatory T cells (Tregs) which in some preclinical models suppress GvHD-causing alloreactive donor T cells but have only limited effects on GvL-promoting alloreactive donor T cells. Unfortunately, Tregs exist in low frequency in the peripheral blood, are costly to purify and expand, and after expansion are difficult to isolate due to the lack of cell surface markers, all of which prevent their routine use in the clinic. Thus, alternative therapeutic approaches that do not require Tregs are needed. We have found that interferon gamma receptor deficient (IFNγR−/−) allogeneic donor T cells induce significantly less GvHD in both a MHC fully-mismatched (B6 (H-2b) → Balb/c (H-2d)) and a minor-mismatched (B6 (H-2b) → B6×129(H-2b)) allo-HSCT models compared to WT T cells. In addition, IFNγR−/− donor T cells maintain a beneficial GvL effect, which has been examined in both systemic leukemia and solid tumor models using luciferase-expressing A20 cells derived from Balb/c. We find that IFNγR−/− T cells migrate primarily to the spleen while WT T cells to GI tract and peripheral lymph nodes (LNs) using bioluminescence imaging (BLI), suggesting that altered T cell trafficking of IFNγR−/− T cells to GvHD target organs might be the major reason for the reduced GvHD. We further demonstrate that the IFNγR-mediated signaling in alloreactive donor T cells is required for expression of CXCR3 which has been implicated in trafficking of T cells to areas of inflammation and target organs, commonly known to be the sites of GvHD. Indeed, CXCR3−/− T cells recapitulate the reduced GvHD potential of IFNγR−/− T cells. In addition, forced overexpression of CXCR3 in IFNγR−/− T cells via retroviral transduction partially rescues the GvHD defect observed in IFNγR−/− T cells. We next examine if inhibition of IFNγR signaling using a small molecule inhibitor can recapitulate the anti-GVHD effects seen in IFNγR−/− T cells. We find that INCB018424, an inhibitor of JAK1/JAK2 which are the mediators of IFNγR signaling, blocks CXCR3 expression in vitro. Most importantly, in vivo administration of INCB018424 after allo-HSCT alters T cell trafficking and significantly reduces GvHD. Thus, the IFNγR signaling pathway represents a promising therapeutic target for future efforts to mitigate GvHD while maintaining GvL after allo-HSCT. Moreover, this pathway can be exploited in other diseases besides GvHD such as those from organ transplantation, chronic inflammatory diseases and autoimmune diseases. Disclosures: DiPersio: genzyme: Honoraria.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2537-2537
Author(s):  
Jaebok Choi ◽  
Julie Ritchey ◽  
Jessica Su ◽  
Julie Prior ◽  
Edward Ziga ◽  
...  

Abstract Abstract 2537 Introduction: Regulatory T cells (Tregs) have been shown to mitigate graft-versus-host disease (GvHD) while preserving the beneficial graft-versus-leukemia (GvL) effect in animal models of allogeneic bone marrow transplantation (BMT). However, three major obstacles prevent their use in human clinical trials: the low numbers of Tregs, loss of suppressor activity following in vitro expansion, and the lack of Treg-specific markers to purify expanded Tregs. The locus of the Foxp3 gene, the master regulator of Tregs, is unmethylated and expressed only in Tregs. We have recently reported that the hypomethylating agent azacitidine (AzaC) induces FOXP3 expression in non-Tregs, converting them into Tregs in vitro and in vivo when administered after allogeneic BMT completely mitigating GvHD without abrogating GvL (Choi, et al Blood 2010). Three possible mechanisms for these effects include: 1) AzaC induces FOXP3+ Tregs, which in turn mitigate GvHD without abrogating GvL by regulating alloreactive donor T cells, 2) AzaC directly suppresses the proliferation of alloreactive donor T cells reducing GvHD, 3) AzaC alters donor T cell trafficking to GvHD target organs to prevent GvHD without altering interaction of donor T cells with recipient leukemia or trafficking of leukemic cells. Methods: Balb/c (CD45.2+, H-2Kd) were lethally irradiated one day prior to injection of T cell-depleted BM cells isolated from B6 (CD45.1+, H-2Kb) and luciferase-expressing A20 leukemia cells derived from Balb/c. Allogeneic donor T cells isolated from B6 (CD45.2+, H-2Kb) were given 11 days after BMT. AzaC (2 mg/kg) was administrated subcutaneously every other day (4 doses total) starting 4 days after T cell injection. In vivo bioluminescence imaging (BLI) was performed to assess leukemia cell localization. For T cell proliferation/trafficking analyses, Balb/c were lethally irradiated one day prior to injection of T cell-depleted BM cells isolated from B6 (CD45.1+). Allogeneic donor T cells isolated from B6 (CD45.2+) were transduced with Click Beetle Red luciferase and were given 11 days after BMT, followed by AzaC treatment as described above. BLI was performed to track the donor T cells. Results: While neither T cell or leukemia cell trafficking was affected by the AzaC treatment, proliferation of donor T cells was significantly reduced compared to mice treated with PBS. The observed reduced T cell proliferation is not likely due to the direct effect of AzaC on T cells since the AzaC treatment preserved GvL activity comparable with the PBS control group. In addition, T cells isolated from both AzaC and PBS groups were equally reactive against third party antigen presenting cells, based on mixed lymphocyte reactions and cytotoxic T lymphocyte killing assays. These data along with our previous report demonstrating that the AzaC treatment increases Tregs in vivo strongly suggest that the therapeutic effect of AzaC on GvHD and GvL are mediated by the AzaC-induced Tregs which preferentially target alloreactive T cells while preferentially sparing anti-tumor T cells. Currently, secondary transplantation of Treg-depleted/replete T cells isolated from AzaC/PBS-treated recipient mice is underway to further confirm that donor T cells in the AzaC-treated mice are fully functional and that alloresponses of donor T cells are regulated by AzaC-induced Tregs. Conclusions: In vivo administration of AzaC after donor T cell infusion mitigates GvHD while preserving GvL via peripheral conversion of alloreactive donor T cells to FOXP3+ Tregs that preferentially inhibit alloreactive T cells while sparing anti-tumor T cells. These data provides the foundation for future clinical trials using epigenetic therapy aimed at mitigating GvHD without abrogating GvL and overcoming HLA barriers. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4464-4464
Author(s):  
Jaebok Choi ◽  
Matthew L Cooper ◽  
Julie Ritchey ◽  
Lynne Collins ◽  
Julie Prior ◽  
...  

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative treatment for patients with relapsed/refractory leukemia, and marrow failure states such as myelodysplasia and aplastic anemia. However, allo-HSCT is complicated by allogeneic donor T cell-mediated graft-versus-host disease (GvHD) which can be life-threatening especially in recipients of unrelated or HLA-mismatched hematopoietic stem cell products. These same alloreactive donor T cells also mediate a beneficial graft-versus-leukemia (GvL) effect. We have recently reported that interferon gamma receptor deficient (IFNγR-/-) allogeneic donor T cells induce significantly less GvHD in both a MHC fully-mismatched (B6 (H-2b) → Balb/c (H-2d)) (75% vs 0% overall survival) and a minor-mismatched (B6 (H-2b) → B6x129 (H-2b)) allo-HSCT models (100% vs 10% overall survival) compared to WT T cells (Choi et al Blood 2012). In addition, IFNγR-/- donor T cells maintain a beneficial GvL effect, which has been examined in both systemic leukemia and solid tumor models using luciferase-expressing A20 cells derived from Balb/c. We also found that IFNγR-/- T cells migrate primarily to the spleen while WT T cells to GI tract and peripheral lymph nodes (LNs) using bioluminescence imaging (BLI), suggesting that altered T cell trafficking of IFNγR-/- T cells to GvHD target organs might be the major reason for the reduced GvHD. We further demonstrated that the IFNγR-mediated signaling (via JAK1/2 - STAT pathway) in alloreactive donor T cells is required for expression of CXCR3 which has been implicated in trafficking of T cells to areas of inflammation and target organs, commonly known to be the sites of GvHD. Here, we examine if inhibition of IFNγR signaling using a small molecule inhibitor can recapitulate the reduced GVHD with potent anti-leukemia effects similarly to that seen with IFNγR-/- T cells. We find that INCB018424, an inhibitor of JAK1/JAK2 which mediate IFNγR signaling, blocks CXCR3 expression in vitro. Most importantly, in vivo administration of INCB018424 (100 ug, s.c., twice a day, day 1-31) after allo-HSCT alters T cell trafficking and significantly reduces GvHD (70% vs. 0% overall survival, n=10/group, p=0.0012). We also find that INCB018424 preserves the beneficial GvL effect, which has been examined in both systemic leukemia and solid tumor models using luciferase-expressing A20 cells derived from Balb/c (B6 to Balb/c model) and APL cells from B6x129 (B6 to B6x129 model). Of note is that INCB018424, when given after transplant, had no significant effect on neutrophil or platelet recovery compared to animals receiving placebo. Thus, the IFNγR signaling pathway represents a promising therapeutic target for future efforts to mitigate GvHD while maintaining GvL after allo-HSCT. Moreover, this pathway could be targeted and exploited in other diseases besides GvHD such as those from organ transplantation, chronic inflammatory diseases and autoimmune diseases. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 806-806 ◽  
Author(s):  
Kevin Beezhold ◽  
Nathan Moore ◽  
Pailin Chiaranunt ◽  
Rebecca Brown ◽  
Craig A. Byersdorfer

Abstract Allogeneic hematopoietic stem cell transplantation (alloHSCT) represents a curative treatment for high-risk leukemia and a number of non-malignant hematologic disorders. However, the therapeutic use of alloHSCT remains limited by acute graft-versus-host disease (GVHD), where activated donor T cells attack and destroy host tissues in the skin, gastrointestinal tract, and liver. We have previously shown that the alloreactive T cells responsible for GVHD increase their dependence on the oxidation of fat relative to either syngeneic or naive T cells. To explore this adaptation mechanistically, we studied the role of AMPK, an intracellular energy sensor and known driver of fat oxidation, in donor T cells during GVHD. Alloreactive T cells increased phosphorylation of AMPK as early as day 3 post-transplant, with up-regulation in pathways both up- and downstream of AMPK. Changes in phosphorylation were up to 8-fold higher in alloreactive T cells compared to naive T cells or syngeneic controls (p=0.0003). We then investigated the role of AMPK during GVHD pathogenesis using donor cells deficient in AMPK (from AMPKα1fl/flα2fl/fl x CD4-Cre mice). AMPK-/- T cells caused significantly less GVHD in both major-MHC and minor-histocompatibility mismatch models of GVHD (Figure 1A), with a coordinated decrease in the number of donor T cells recovered on day 7 post-transplant (3.15 +/- 0.49x106 versus1.87 +/- 0.53x106, p=0.0006, wildtype (wt) versus AMPK-/- respectively). Importantly, expansion of syngeneic T cells was unaffected by AMPK deficiency (Figure 1B). We next investigated the ability of AMPK-/- T cells to mount effective cytotoxic and anti-leukemia responses. AMPK-/- T cells demonstrated equivalent cytotoxicity against MHC-mismatched targets both in vitro and in vivo and differentiated in similar proportions into cytokine-producing cells (IFN-γ, TNFα, IL-17, and IL-4). We then assessed graft-versus-leukemia (GVL) potential in AMPK-/- cells using a GVL model with high tumor burden. AMPK-/- T cells exhibited equivalent clearance of p815 leukemia cells on day 13 post-transplant (Figure 2A), and extended survival of recipient mice similarly to wt T cells (Figure 2B). To elucidate possible mechanisms underlying this separation of GVL and GVHD responses, we evaluated metabolic pathways in wt and AMPK-/- T cells recovered on day 7. To our surprise, rates of fatty acid oxidation were identical between wt and AMPK-/- T cells and loss of AMPK did not impact alloreactive T autophagy, nor impair signaling downstream of mammalian target of rapamycin. To define the mechanism underlying AMPK-/- benefits, we quantitated levels of regulatory T cells (Treg) on day 7 post-transplant. In contrast to expectation, both the percentage and total number of Treg increased in mice receiving AMPK-/- T cells (0.85 +/- 0.32x104 vs. 1.69 +/- 0.34x104, wt vs. AMPK-/-, p=0.004). Loss of AMPK facilitated donor Treg expansion, as elimination of FoxP3+ cells prior to transplantation abrogated differences between wt and AMPK-/- donors on day 7. Importantly, Treg levels were equal in wt versus AMPK-/- donors prior to transplantation. Finally, we assessed the ability of AMPK-/- T cells to infiltrate into GVHD target organs. As shown in Figure 3, peri-portal infiltration of AMPK-/- cells was significantly reduced compared to wt T cells, and infiltrates in recipients of AMPK-/- cells contained many fewer CD3+ T cells per high-powered field, with CD3+ cells representing a lower percentage of cells overall. Decreased hepatic infiltration correlated with a lower percentage of circulating CD4+ cells and lower levels of the integrin pair α4β7 (55.2 +/- 1.4% versus 47.2 +/- 2.8% α4β7Hi cells, p=0.0017, wt vs. AMPK-/-). In conclusion, deletion of AMPK in donor T cells decreases GVHD severity but spares anti-leukemia responses and preserves homeostatic immune reconstitution. Mechanistically, this occurs through a decrease in pathogenic T cell numbers, an increase in the number and percentage of Treg cells, fewer circulating CD4+ cells, and decreased infiltration of donor cells into target organs. From these findings, we conclude that AMPK represents a clinically relevant target in donor T cells pre-transplant and are actively exploring ways to translate this exciting therapy into clinical practice. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2162-2162
Author(s):  
Janelle A. Olson ◽  
Dennis B. Leveson-Gower ◽  
Andreas Beilhack ◽  
Robert S. Negrin

Abstract Natural Killer (NK) cells have the ability to suppress graft-versus-host disease (GVHD) while inducing a graft-versus-tumor response (GVT) during allogeneic bone marrow transplantation (BMT). Previous studies in allogeneic BMT models have shown NK cell trafficking to and proliferation in lymphoid organs and GVHD target organs, which are also sites of donor T cell trafficking. This study aims to investigate the impact of NK cells on alloreactive, GVHD-inducing donor T cells. Interleukin-2 activated allogeneic NK cells isolated from C57Bl6 (H–2b) or FVB (H–2q) animals were transplanted along with T cell-depleted bone marrow into lethally irradiated BALB/c (H–2d) mice, followed 2 days later by luciferase-expressing CD4+ and CD8+ conventional T cells from the same donor strain (NK+Tcon group). Control mice received lethal irradiation and T cell-depleted bone marrow on day 0, and luciferase-expressing T cells on day 2 after transplant (Tcon group). Bioluminescence imaging of NK+Tcon mice revealed a significantly lower T cell bioluminescent signal (p=0.03 for FVB into BALB/c on day 6) than from Tcon mice. CFSE proliferation analysis of alloreactive T cells on day 3 after transplant showed no significant change in the percent of donor T cells that have divided in the spleen, and only a slight decrease in the percent of T cells that have divided in the lymph nodes when NK cells are present. However, at this timepoint 82% of the proliferating cells have divided past the third generation, in contrast to 64% in the NK+Tcon mice. Donor T cells in both groups become equally activated in vivo, expressing similar levels of the early-activation marker CD69. T cells re-isolated from NK+Tcon animals at day 5 stained 2 to 10-fold higher for the TUNEL apoptosis marker than those from Tcon mice in the mesenteric and peripheral lymph nodes, respectively (p&lt;0.0001). Additionally, decreased numbers of T cells were re-isolated from the peripheral lymph nodes in the NK+Tcon group as compared to the Tcon group. This increase in TUNEL staining was not seen when the transplanted NK cells were isolated from a perforin-deficient donor. This indicates that NK cells in lymph nodes use a perforin-dependent mechanism to increase apoptosis in proliferating, alloreactive donor T-cells, which are syngeneic to the transplanted NK cells. Donor T cells re-isolated from the lymph nodes of transplanted mice up-regulate the NKG2D ligand Rae1γ as compared to naïve T cells, as shown by FACS. This suggests that NK cells may cause direct lysis of alloreactive donor T cells in vivo during GVHD induction, mediated by the NK cell activating receptor NKG2D. This study provides crucial mechanistic information regarding the function of NK cells in suppressing GVHD.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3506-3506
Author(s):  
Jonathan Serody ◽  
Chris A Wysocki ◽  
Tim P Moran

Abstract Background: Previous investigators have demonstrated that secondary lymphoid tissue (SLT) including the spleen is critical for the initial activation of donor T cells after allogeneic stem cell transplantation (SCT) leading to GVHD. This work showed significant redundancy in the role of all SLT including the spleen and suggested that approaches to prevent GVHD would need to inhibit the migration of donor T cells to all SLT. The mechanisms mediating the migration of donor T-cells to SLT have not been well characterized and the specific role of conditioning therapy in this process has not been investigated. Thus, here we sought to evaluate the expression of trafficking proteins by host antigen presenting cells isolated from the MLN and spleen. Methods: B6D2 recipient mice that underwent conditioning were lethally irradiated the night prior to transplantation. The following day recipient mice were transplanted i.v. with 3 × 106 T cell depleted bone marrow cells with 5 × 106 column purified T-cells. T-cells were from eGFP C57Bl/6 transgenic mice. Six hours after transplantation, recipient mice were sacrificed and the MLN and spleen removed. Dendritic cells (DCs) were isolated by collagenase digestion and mechanical disruption and sorted on a high speed MoFlow sorter using anti-CD11c monoclonal antibodies. Gates were drawn around CD11c high cells that did not express eGFP. 3–5 × 104 DCs from the MLN and 1–2 × 105 DCs from the spleen were pooled from five different mice per group, lysed in an RNA amplification buffer and analyzed either by real-time PCR or Agilent Whole Mouse Genome Microarray Chips. Results: Four different groups of mice were analyzed including one group that received no treatment, one that received irradiation alone, one that received irradiation plus allogeneic bone marrow and T cells, and one that received allogeneic bone marrow and T cells without irradiation. By comparing recipients that received allogeneic bone marrow and T cells, with those that received conditioning therapy, allogeneic bone marrow and T cells, we found that conditioning therapy significantly induced the expression by host DCs of the chemokines, CXCL10, CCL17, CCL20 and CCL22 from DCs isolated from both the MLN and the spleen. CCL3 expression by host cells was only upregulated from DCs isolated from the MLN and not the spleen. Conditioning therapy downregulated the expression of CCL1, CCL2 and CCL4 from DCs isolated from both the MLN and spleen (Table 1). Conditioning therapy markedly enhanced the expression by splenic and MLN DCs of STAT4 and p40 IL-12. Table 1 MLN Spleen CXCL10 14.8 20 CCL3 10.9 −1.0 CCL17 7.0 5.1 CCL22 2.4 7.8 CCL20 2.2 1.6 CCL6 1.3 1.2 CCL5 −1.3 3.0 CCL19 −1.4 −2.3 CXCL9 −1.5 −1.3 CCL1 −2.2 2.2 CCL2 −3.7 −3.0 CCL4 −6.0 −1.4 Next we analyzed the effect of allogeneic bone marrow and T cells on the expression of chemokine ligands and signaling proteins by splenic and MLN DCs. Interestingly, the receipt of allogeneic bone marrow plus T cells and conditioning therapy led to a marked increase in the expression of chemokine ligands by DCs isolated from the spleen with little effect on the expression of chemokine ligands by DCs isolated from the MLN compared to mice that only received irradiation but did not undergo allogeneic transplantation Table 2. MLN Spleen CXCL9 1.1 57 CCL22 2.2 36 CCL5 −1.1 34 CXCL10 2.1 30 CCL17 4.3 8.1 CCL4 −1.1 7.2 CCL6 1.4 6.7 CCL3 6.0 1.0 CCL20 2.4 1.0 CCL1 2.5 −1.5 CCL19 −4.2 −4.0 CCL2 −6.7 −7.1 The receipt of irradiation, donor bone marrow and T cells markedly enhanced the expression by host DCs from the spleen of STAT4 and p40 IL-12 while downregulating the expression of STAT6. We confirmed that the expression of CXCL9, CXCL10, CCL4 and CCL5 were markedly increased by host APCs using real time PCR. Finally, we demonstrated in vitro that donor T cell production of IFN-g was critical to the generation of CXCL9 and CXCL10 by host APCs. Conclusion: Conditioning therapy markedly enhances the expression of chemokine ligands and proteins important in Th1 T cell activation by host APCs. However, the expression was enhanced the most in splenic DCs isolated from recipient mice that had received irradiation with allogeneic bone marrow and allogeneic T cells. Our work suggests that the processes mediating the migration of donor T cells to the spleen may differ from those in the MLN and that the CXCR3 binding chemokines, CXCL9 and CXCL10 along with the CCR5 binding chemokine, CCL3, may be quite critical for donor T cell homing to the spleen.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3829-3829
Author(s):  
Jaebok Choi ◽  
Matthew L Cooper ◽  
Bader Alahmari ◽  
Julie Ritchey ◽  
John F. DiPersio

Abstract The therapeutic benefits of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for hematologic malignancies are primarily derived from anti-leukemia effect that is mediated by T cells in donor grafts. Unfortunately, these T cells also mediate graft-versus-host disease (GvHD), the major complication of allo-HSCT. We have recently reported that blockade of alloreactive donor T cell trafficking to the GvHD target organs significantly reduces GvHD in both a MHC fully-mismatched (B6 (H-2b) → Balb/c (H-2d)) (75% vs 0% overall survival) and a minor-mismatched (B6 (H-2b) → B6x129 (H-2b)) allo-HSCT models (100% vs 10% overall survival) compared to WT T cells (Choi et al., Blood 2012). In this study, we examined if inhibition of VLA-4, which is required for transendothelial migration and access to GvHD target organs, could reduce GVHD. VLA-4 consists of two subunits α4 (CD49d) and β1 (CD29). To genetically eliminate VLA-4 from allogeneic donor T cells, we generated Tie-2 cre+ α4-/- mice (B6, H-2b, CD45.2+) in which α4 is knocked out by Tie-2 cre, which is expressed in hematopoietic cells. Splenic pan T cells were isolated from these mice and T cell-depleted bone marrow cells (TCD BM) from congenic B6 mice (CD45.1+). 5x106 TCD BM and 5x105 splenic pan T cells were transplanted into lethally irradiated allogeneic Balb/c recipient mice (H-2d, CD45.2+). We found that VLA-4 deficient T cells significantly reduced GvHD compared to WT T cells (Fig. A). VLA-4 deficient T cells had no significant effect on donor engraftment and achieved a complete donor chimerism. In addition, recipients transplanted with VLA-4 deficient T cells had significantly better histopathology score (Fig. B). Similar results were observed when donor T cells were infused at day 11 after allo-HSCT. While we speculate defective T cell trafficking to GvHD target organs would be the primary reason for the reduced GvHD in the recipients of VLA-4 deficient T cells, we examined other possible mechanisms as well, such as T cell alloreactivity and proliferation. We found that VLA-4 deficient T cells proliferated at the same rate as WT T cells in the presence of anti-CD3/CD28 antibody-coated beads. However, VLA-4 deficient T cells proliferate less in the presence of allogeneic antigen presenting cells (APCs) compared to WT T cells. Interestingly, VLA-4 deficient T cells upregulate CTLA-4 and GZMB significantly more than WT T cells after in vitro activation. These data suggest that not only T cell trafficking to GvHD target organs but also altered T cell phenotype and function might help explain the observed reduced GvHD. Genetic deletion of α4 in T cells will also alter expression of α4b7. Since antibodies to b7 have also been shown to reduce GvHD (Waldman et al., Blood, 2006) our current studies will focus on determining which of these pathways (α4b1 vs. α4b7) is most important in mitigating GvHD by using genetic knockout models, antibodies to b1 and b7 and small molecule inhibitors of α4b1 vs. α4b7. In conclusion, our data suggests that VLA-4 represents a promising therapeutic target for future efforts to mitigate GvHD after allo-HSCT. In addition, this strategy can be exploited in other diseases and settings in addition to allo-HSCT, such as solid organ transplantation, chronic inflammatory diseases and autoimmune diseases. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 41 (Supplement_2) ◽  
Author(s):  
D Vdovenko ◽  
W.J Wijnen ◽  
M Zarak Crnkovic ◽  
P Blyszczuk ◽  
M Bachmann ◽  
...  

Abstract Introduction Cardiac inflammation varies widely and, in some cases, triggers autoimmune myocarditis and further inflammatory dilated cardiomyopathy (iDCM) and heart failure. In children, myocarditis leads to cardiomyopathy in almost half of affected individuals and up to 20% of sudden death cases in young adults have been reported to be due to myocarditis. IL-12 and IL-23 belong to the same family of cytokines known to mediate inflammatory conditions. Both regulate the differentiation of T cells: IL-12 promotes towards IFN-gamma-producing Th-1 cells, while IL-23 induces IL-17-producing Th-17 cells. Heart-reactive CD4+Th17 cells play a leading role in the development of myocarditis, however, literature reports excessiveness of IL-23 in Th17-derived IL-17 production. Therapeutic strategies blocking IL-23 were suggested as a promising approach, though the specific role of IL-23 in pathogenesis is unclear and the long-term perspectives stay elusive. Purpose We aim to explore the role IL-23 compared to IL-12 in the manifestation of cardiac autoimmune myocarditis. Methodology We use dendritic cell (DC) model of experimental autoimmune myocarditis in IL-12 and IL-23-deficient mice. Mice were injected with bone marrow-derived in vitro activated and loaded with cardiac-specific peptide DCs. This model mimics natural processes taking place during heart inflammation and provides a unique method to address the role of DCs-derived cytokines. Cardiac inflammation, as well as remodeling and heart function, were analysed at the acute and chronic stages of the disease. Results Surprisingly, all mice developed acute myocarditis, though wt receiving IL-23−/−bmDCs showed a twofold decrease in heart-infiltrating T cells and lower numbers of Th17 population. Further decrease of heart-infiltrating T cells appeared upon total systemic IL-23 deficiency. In comparison to IL-12, directly inducing differentiation of IFN-gamma–producing Th1, IL-23 cannot induce Th17 differentiation. None of the two cytokines affect proliferation, though, IL-23 activates T cell migratory potential and increases T cell migration by twofold. At the same time, deficiency of IL-23-production by bmDCs leads to lower migration of T cells. We also show an involvement of RhoA, and the other Rho GTPases, in the mechanism of migration as blocking revoke the IL-23 effect on T cells. Moreover, we further observed more fibrosis and worse heart functioning in IL-23−/−, but not IL-12−/− mice at the chronic stage what underlines the importance of IL-23-dependent T cell trafficking in the resolution of the acute stage of autoimmune myocarditis. Conclusion Our observations underline IL-23 as an important cytokine responsible for T cell trafficking and resolution of the inflammation in autoimmune myocarditis. Therapeutic approaches involving inflammatory cytokine targeting are a promising clinical perspective though IL-23 deficiency might lead to increased cardiac remodeling and iDCM progression. Funding Acknowledgement Type of funding source: Foundation. Main funding source(s): The Swiss National Science Foundation


Sign in / Sign up

Export Citation Format

Share Document