Visualizing Allogeneic Bone Marrow (BM) Graft Rejection.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 43-43
Author(s):  
Patricia Taylor ◽  
Angela Panoskaltsis-Mortari ◽  
Randolph Noelle ◽  
Alexander Rudensky ◽  
Jonathan Serody ◽  
...  

Abstract The process by which bone marrow is rejected by host T cells has not been able to be directly visualized to date. To study the process of allogeneic bone marrow rejection and the effects of therapeutic interventions, we created 2 models that allow us to 1) quantify the specific expansion of host-type alloreactive T cells early post-transplant in response to allogeneic BM infusion and to 2) image host T cells in vivo during BM rejection. For the first model, 2C and TEa lymph node (LN) cells were adoptively transferred into syngeneic C57BL/6 (B6) Rag deficient mice on d-2. 2C CD8+ and TEa CD4+ T cell receptor transgenic T cells are reactive against BALB/c alloantigen. Mice were irradiated with 200 cGy on d-1 and BALB/c BM was infused on d0. Controls included mice that received 2C/TEa LN cells but no BM. Ten days later, spleen analysis revealed that 2C CD8+ and TEa CD4+ T cells had expanded 322-fold and 33-fold (ave of 6 exp.), respectively, in mice receiving BALB/c BM compared to controls that did not receive BM. Expanded T cells were activated as determined by flow cytometric parameters and cell surface antigens. Data indicate that host alloreactive T cell expansion was inhibited by >95% by combined, but not single, costimulatory pathway blockade. Studies are in progress to analyze in vitro host anti-donor responses of adoptively transferred T cells. To visualize the response of host T cells to donor BM in vivo, we developed a rejection model for imaging involving the adoptive transfer of green fluorescent protein (GFP) T cells (obtained from GFP transgenic mice) into syngeneic non-GFP B6 recipients immediately following sublethal irradiation (500 cGy). Allogeneic BALB/c or syngeneic B6 BM was infused the following day. The syngeneic BMT controls allowed for the distinction of homeostatic vs alloreactive expansion of GFP+ T cells. Transplanted mice not receiving GFP T cells served as negative controls to verify lack of autofluorescence. Cohorts were imaged d4 to d18 post BMT. By d4, low numbers of GFP+ cells were evident in femoral BM cavity, peripheral and mesenteric LNs, spleen, Peyer’s patches (PP) and to a lesser extent, lung. By d7, massive expansion of GFP+ cells could be visualized throughout the body of recipients of allogeneic BM. LNs, spleen, PP (peri-follicular area) and BM cavity increased dramatically in GFP intensity from d4 to d7. On d7 to d14, there were large foci of GFP+ cells in the lung, liver, skin, gingiva, kidney, uterus, and colon in allogeneic BMT recipients. Compared with allogeneic BMT recipients, syngeneic BMT recipients had greatly reduced numbers of GFP+ T cells in lymphoid organs and only rare cells were noted in liver, kidney, skin, BM and gingiva. In both allogeneic and syngeneic BMT recipients, lengths of ileum were diffusely infiltrated while other sections contained discrete foci of GFP+ cells. These imaging data provide a vivid illustration of the massive expansion and multi-organ distribution of host anti-donor T cells in vivo. Recent generation of GFP+ 2C and GFP+ TEa mice will permit the imaging of alloantigen-specific T cells during a rejection response. Additional imaging experiments are planned to study the fate of GFP+ BM transferred to allogeneic recipients under conditions of engraftment vs rejection. These models provide a unique platform for the testing of therapeutic interventions.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2960-2960
Author(s):  
Ying Wang ◽  
Jian-Ming Li ◽  
Wayne A.C. Harris ◽  
Cynthia R. Giver ◽  
Edmund K Waller

Abstract Abstract 2960 Background: Donor cell engraftment following allogeneic bone marrow transplantation (BMT) is affected by several factors, including immunological major histocompatibility complex (MHC) barriers, the intensity of the conditioning regimen, and the content of T-cells in the graft. The current model for engraftment in allogeneic BMT is that host dendritic cells (DCs) activate donor T-cells which promote engraftment by eliminating radio-resistant cytotoxic host immune cells, especially natural killer (NK) cells and T-cells. To explore the interaction between donor T-cell and host antigen-presenting cells (APC) in engraftment in allogeneic BMT, we focused on the role of interleukin-12 (IL-12), a key cytokine produced mainly by DCs that drives the development of donor type 1 helper T cells (Th1) and type 1 cytotoxic T lymphocytes (Tc1). Methods: Radiation chimeras with >95% donor chimerism were created by transplanting 5 × 106 bone marrow (BM) cells from IL-12 knock out (IL-12 KO) or wild type (WT) B6 (H-2Kb, CD45.2) donors into congenic BL6 Pepboy (B6.SJL-PtprcaPep3b/BoyJ, H-2Kb, CD45.1) mice following lethal 11 Gy irradiation. A second allogeneic BMT was conducted 2 months later using MHC mismatched FVB (H-2q, CD45.1), BA.B10 (H-2Kk, CD45.2, CD90.1) or B10.BR (H-2Kk, CD45.2, CD90.2) donor cells. In vivo bioluminescent imaging (BLI) was performed to analyze the number and in vivo distribution of luciferase+ donor T-cells. The whole-body bioluminescent signal was used as a marker of the donor T cell expansion. Engraftment of donor myeloid cells was determined by flow cytometry using mAbs for specific leukocyte markers expressed on donors and recipients (CD45.1, CD45.2, H-2Kb). Intracellular cytokine expression (IL-4, IL-10, IFN-g) by donor CD4+ and CD8+ T cells was analyzed by flow cytometry. Results: WT BL6→BL6 radiation chimeras recipients showed greater expansion of luciferase+ donor T-cells compared with IL-12 KO BL6→BL6 radiation chimeras recipients and FVB→FVB syngeneic recipients at early time point (2 wks) following 9 Gy re-irradiation and transplantation of 3 × 105 luciferase+ FVB-L2G85 T-cells in combination with 5 × 106 T cell depleted (TCD) BM cells from FVB mice following (Fig 1). At 4 weeks post transplant, more WT BL6→BL6 radiation chimeras achieved myeloid engraftment than IL-12 KO BL6→BL6 radiation chimera recipients(75.0% versus 33.3% respectively, p = 0.086), and the former group had better erythroid engraftment than the latter group (RBC 8.65 ± 1.88 × 1012/L versus 5.67 ± 2.22 × 1012/L respectively, p = 0.011). However, when FVB, WT BL6→BL6 or IL-12 KO BL6→BL6 radiation chimeras recipients were conditioned with a larger dose of irradiation prior to the second transplantation (10 Gy) and received a larger dose of donor T-cells (5 × 105), both the WT BL6→BL6 and IL-12 KO BL6→BL6 radiation chimeras recipients achieved full donor engraftments (85.7% versus 87.5% respectively, p = NS). Donor T cells in allogeneic BMT recipients were Th1/Tc1 polarized, there were no differences in frequencies and total numbers of Th1/Tc1 donor CD4+ and CD8+ T cells comparing recipients of WT BL6→BL6 and IL-12 KO BL6→BL6 radiation chimeras. In spite of an increased irradiation dose and larger number of donor T-cells in the second transplant regimen, no increase in graft versus host disease (GVHD) clinical scores and GVHD-mortality were observed in the recipients of WT BL6→BL6 radiation chimeras compared with recipients of IL-12 KO BL6→BL6 radiation chimeras. Conclusion: These data support a role for host BM-derived IL-12 in facilitating engraftment in allogeneic BMT following a reduced dose (9 Gy) radiation. The lack of host BM-derived IL-12 expression led to allograft rejection. Rejection could be overcome by increasing the dose of pre-transplant irradiation and the content of donor T-cells without causing lethal GVHD. As the main source of host BM-derived IL-12, recipient APC thus play an important role in donor T-cell activation. As has been previously demonstrated in a murine BMT model, the addition of IL-12 in the peri-transplant period helped to separate graft versus leukemia effects from the GVHD-promoting activity of donor T-cells (Yang, 1997). Patients predicted to be high risk of graft failure may benefit from treatment strategies that contribute to production of IL-12 during the early phases of hematopoietic engraftment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 109 (9) ◽  
pp. 4080-4088 ◽  
Author(s):  
Mathias M. Hauri-Hohl ◽  
Marcel P. Keller ◽  
Jason Gill ◽  
Katrin Hafen ◽  
Esther Pachlatko ◽  
...  

Abstract Acute graft-versus-host disease (aGVHD) impairs thymus-dependent T-cell regeneration in recipients of allogeneic bone marrow transplants through yet to be defined mechanisms. Here, we demonstrate in mice that MHC-mismatched donor T cells home into the thymus of unconditioned recipients. There, activated donor T cells secrete IFN-γ, which in turn stimulates the programmed cell death of thymic epithelial cells (TECs). Because TECs themselves are competent and sufficient to prime naive allospecific T cells and to elicit their effector function, the elimination of host-type professional antigen-presenting cells (APCs) does not prevent donor T-cell activation and TEC apoptosis, thus precluding normal thymopoiesis in transplant recipients. Hence, strategies that protect TECs may be necessary to improve immune reconstitution following allogeneic bone marrow transplantation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3482-3482
Author(s):  
Minghui Li ◽  
Kai Sun ◽  
Mark Hubbard ◽  
Doug Redelman ◽  
Angela Panoskaltsis-Mortari ◽  
...  

Abstract IL-17-producing CD4 T cells (Th17) are a recently identified T helper subset that plays a role in mediating host defense to extracellular bacteria infections and is involved in the pathogenesis of many autoimmune diseases. In vitro induction of IL-17 in murine CD4+ T cells has been shown to be dependent on the presence of the proinflammatory cytokines TGF-β and IL-6 whereas IFNγ can suppress the development of Th17 cells. In the current study, we examined the roles of TNFα and IFNγ on IL-17 production by purified T cells in vitro and in vivo after allogeneic bone marrow transplantation (BMT). We present findings that expression of TNFα by the T cell itself is necessary for optimal development of Th17 under in vitro polarizing conditions. A novel role for T cell-derived TNFα in Th17 induction was observed when in vitro polarization of Tnf−/−CD4+ T cells resulted in marked reductions in IL-17+CD4+ T cells compared to Tnf+/+CD4+ T cells. In marked contrast, T cell-derived IFNγ markedly inhibited Th17 development as more IL-17+CD4+ T cells were found in Ifnγ−/−CD4+ T cells than in Ifnγ+/+CD4+ T cells, and of particular interest was the dramatic increase in IL-17+CD8+ cells from Ifnγ−/− mice. To determine if T cell-derived TNFα or IFNγ can regulate Th17 development in vivo we examined the differentiation of alloreactive donor T cells following allogeneic BMT. We have found that donor-derived Th17 cells can be found in lymphoid tissues and GVHD-affected organs after allogeneic BMT. However, transfer of Tnf−/− CD4+ T cells after allogeneic BMT resulted in marked reductions in Th17 cells in the spleen (18×103 vs 7×103, P<0.05). In agreement with the in vitro data and in contrast to what was observed with transfer of Tnf−/− CD4+ T cells, transfer of donor Ifnγ−/− T cells resulted in marked increases in not only IL-17+CD4+ but also IL-17+CD8+ T cells infiltrating the liver (7×103 vs 14×103, P<0.05; 4×104 vs 12.5×104, P<0.05). These results suggest that the donor T cell-derived TNFα and IFNγ opposingly regulate IL-17 induction of both CD4+ and CD8+ T cells in vitro and after allogeneic BMT which correlates with GVHD pathology.


Blood ◽  
2008 ◽  
Vol 112 (4) ◽  
pp. 1522-1529 ◽  
Author(s):  
Kai Sun ◽  
Minghui Li ◽  
Thomas J. Sayers ◽  
Lisbeth A. Welniak ◽  
William J. Murphy

Abstract Dissociating graft-versus-tumor (GVT) effect from acute graft-versus-host disease (GVHD) still remains a great challenge in allogeneic bone marrow transplantation (allo-BMT). Bortezomib, a proteasome inhibitor, has shown impressive efficacy as a single agent in patients with hematologic malignancies but can result in toxicity when administered late after allogeneic transplantation in murine models of GVHD. In the current study, the effects of T-cell subsets and their associated cytokines on the efficacy of bortezomib in murine allogeneic BMT were investigated. Increased levels of serum tumor necrosis factor-α (TNFα) and interferon-γ (IFNγ) were observed after allo-BMT and continuous bortezomib administration. Bortezomib-induced GVHD-dependent mortality was preventable by depletion of CD4+ but not CD8+ T cells from the donor graft. The improved survival correlated with markedly reduced serum TNFα but not IFNγ levels. Transfer of Tnf−/− T cells also protected recipients from bortezomib-induced GVHD-dependent toxicity. Importantly, prolonged administration of bortezomib after transplantation of purified CD8+ T cells resulted in enhanced GVT response, which was dependent on donor CD8+ T cell–derived IFNγ. These results indicate that decreased toxicity and increased efficacy of bortezomib in murine allo-BMT can be achieved by removal of CD4+ T cells from the graft or by inhibiting TNFα.


2000 ◽  
Vol 111 (1) ◽  
pp. 303-313 ◽  
Author(s):  
Jurgen Finke ◽  
Hartmut Bertz ◽  
Claudia Schmoor ◽  
Hendrik Veelken ◽  
Dirk Behringer ◽  
...  

2003 ◽  
Vol 2 (3) ◽  
pp. 237-260
Author(s):  
Margot Zöller

The critical role of antigen-specific T cells in cancer immunotherapy has been amply demonstrated in many model systems. Though success of clinical trials still remains far behind expectation, the continuous improvement in our understanding of the biology of the immune response will provide the basis of optimized cancer vaccines and allow for new modalities of cancer treatment. This review focuses on the current status of active therapeutic vaccination and future prospects. The latter will mainly be concerned with allogeneic bone marrow cell transplantation after non-myeloablative conditioning, because it is my belief that this approach could provide a major breakthrough in cancer immunotherapy. Concerning active vaccination protocols the following aspects will be addressed: i) the targets of immunotherapeutic approaches; ii) the response elements needed for raising a therapeutically successful immune reaction; iii) ways to achieve an optimal confrontation of the immune system with the tumor and iv) supportive regimen of immunomodulation. Hazards which one is most frequently confronted with in trials to attack tumors with the inherent weapon of immune defense will only be briefly mentioned. Many question remain to be answered in the field of allogeneic bone marrow transplantation after non-myeloablative conditioning to optimize the therapeutic setting for this likely very powerful tool of cancer therapy. Current considerations to improve engraftment and to reduce graft versus host disease while strengthening graft versus tumor reactivity will be briefly reviewed. Finally, I will discuss whether tumor-reactive T cells can be “naturally” maintained during the process of T cell maturation in the allogeneic host. Provided this hypothesis can be substantiated, a T cell vaccine will meet a pool of virgin T cells in the allogeneically reconstituted host, which are tolerant towards the host, but not anergised towards tumor antigens presented by MHC molecules of the host.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2180-2180
Author(s):  
Christian M. Capitini ◽  
Sarah Herby ◽  
Crystal L. Mackall ◽  
Terry J. Fry

Abstract BACKGROUND: Acute graft versus host disease (GVHD) remains as the major complication after allogeneic bone marrow transplant (BMT) resulting in organ toxicity and immune dysfunction. Indeed, we have previously demonstrated that GVHD impairs responses to dendritic cell vaccines. The pathophysiology of GVHD involves preparative regimen-induced inflammation of target organs, release of inflammatory mediators such as gamma interferon (IFNg), and subsequent activation of alloreactive T cells. Given that IFNg can both contribute to GVHD and provide beneficial immune responses, we explored the potential role of IFNg on GVHD and post-transplant immunocompetence. METHODS: We utilized a minor histocompatibility antigen mismatched, T cell-depleted BMT model, with delayed donor lymphocyte infusions (DLIs) as a means of controlling the induction of acute GVHD and to provide a source of immunocompetence in a thymectomized mouse. To study the role of IFNg on GVHD, we chose either IFNg receptor 1 (IFNgR1) −/− marrow or DLI to permit the normal production of IFNg in GVHD while influencing which cells that can respond to the cytokine. Normal C57BL/6 (B6) or IFNgR1 −/− B6 mice were used as bone marrow donors on day 0 into lethally irradiated, thymectomized B6 × C3H.SW (F1) mice. Normal B6 or IFNgR1 −/− DLIs given with or without a dendritic cell vaccine were introduced at days 14 and 28 post-BMT both to control the induction of GVHD and to provide a population of vaccine-responding cells. F1 recipients were observed for signs of GVHD. ELISPOT of the number of antigen-reactive IFNg-producing splenocytes were also performed to measure functional response to vaccine. RESULTS: The absence of IFNgR1 in the DLI abrogates GVHD as shown by % change in weight (B6 DLI = −6.3 +/− 4.7 vs. IFNgR1−/− DLI = 6.6 +/− 6.1, p=0.001) and allows for greater doses of DLI to be tolerated by the host, however, there is also decreased vaccine responses by ELISPOT (B6 DLI = 1631 vs. IFNgR1−/− DLI = 72, p=0.03). Surprisingly, using IFNgR1−/− bone marrow also abrogates GVHD as shown by % change in weight (B6 marrow = −6.3 +/− 4.7 vs. IFNgR1−/− marrow = 4.4 +/− 4.2, p less than 0.05) and splenocyte count (B6 marrow = 31.48 +/− 12.54 vs. IFNgR1−/− marrow = 63.54 +/− 15.92, p=0.008), but vaccine responses by ELISPOT can be restored to levels that are equivalent of syngeneic control mice, even in the presence of a normal B6 DLI (B6 marrow = 1631 vs. IFNgR1−/− marrow = 9283, p=0.0002). The abrogation of GVHD by IFNgR1−/− marrow does not appear to be a dominant effect since mixtures of IFNgR1 −/− and normal B6 bone marrow still cause GVHD. CONCLUSIONS: Recipients of allogeneic bone marrow and T cells developed GVHD and had decreased vaccine responses by ELISPOT. Loss of IFNgR1 on allogeneic donor lymphocytes abrogates their ability to cause GVHD, but also diminished their ability to respond to vaccine. Surprisingly, loss of IFNgR1 on a donor bone marrow-derived, non T cell results in equivalent abrogation of GVHD, while restoring immunocompetence through favorable responses to a vaccine. Further studies will attempt to identify the phenotype of the responsible bone marrow-derived cell. These results demonstrate a strategy of providing higher doses of DLIs to enhance anti-tumor activity without exacerbating GVHD, and thus, have implications for immune modulation post-allogeneic BMT.


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.


Sign in / Sign up

Export Citation Format

Share Document