T-cell subsets, bm mutants, and the mechanisms of allogeneic skin graft rejection

1989 ◽  
Vol 8 (2) ◽  
pp. 149-164 ◽  
Author(s):  
Hugh Auchincloss ◽  
Theodore Mayer ◽  
Rafik Ghobrial ◽  
Henry J. Winn
10.1038/2001 ◽  
1998 ◽  
Vol 4 (9) ◽  
pp. 1015-1019 ◽  
Author(s):  
Stephan Ehl ◽  
Peter Aichele ◽  
Hansruedi Ramseier ◽  
Winfried Barchet ◽  
Joachim Hombach ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1874-1874 ◽  
Author(s):  
Brian C Betts ◽  
David Bastian ◽  
Hung Nguyen ◽  
Jessica Lauren Heinrichs ◽  
Yongxia Wu ◽  
...  

Abstract Janus kinase 2 (JAK2) signal transduction is a critical mediator of immune response. JAK2 activation promotes T-cell allosensitization, as well as Th1 and Th17 differentiation. JAK2-mediated STAT3 phosphorylation limits the generation of induced regulatory T cells (iTregs) by disrupting interactions between STAT5 and Foxp3. JAK2 is implicated in the onset of graft-versus-host disease (GVHD), which is a significant cause of transplant-related mortality after allogeneic hematopoietic cell transplantation (allo-HCT). Using murine models of allo-HCT we show here that transfer of donor JAK2-/- T cells is associated with significantly less GVHD, compared with wild-type or JAK2 replete donors (Figure 1, P =.003 and 0.01, respectively). Th1 differentiation among JAK2-/- T-cells is dramatically decreased, compared with controls. Conversely, iTreg polarization and stability are significantly increased among the JAK2 deficient T cells. To investigate the effect of pharmacologic JAK2 inhibition on T-cell alloresponses, pacritinib (supplied by CTI BioPharma), was chosen as it does not induce myelosuppression or increase risk for opportunistic infections in myelofibrosis patients - unlike JAK1/JAK2 inhibitors. Pacritinib potently inhibits JAK2, but also has suppressive activity toward JAK3, CSF1R, and IRAK1. Pacritinib was administered at 100mg/kg twice a day by oral gavage for 4 weeks beginning on the day of MHC-disparate allo-HCT, significantly reducing GVHD in recipient mice compared with methylcellulose vehicle control. In allogeneic mixed leukocyte reactions using human cells, pacritinib (2.5μM) significantly reduces T-cell proliferation after 5 days of culture (P <.0001). In vitro studies verified pacritinib eliminates STAT3 activity by IL-6 in human CD4+ T-cells, while permitting IL-2 induced STAT5 phosphorylation despite its effects on JAK3. This offers a platform to reduce Th17 development, while promoting iTreg populations. As such, pacritinib dramatically suppresses Th17 differentiation reflected by RORgammaT expression among naïve CD4+ T cells stimulated by allogeneic dendritic cells (DC), compared with DMSO (P <.0001). iTregs were generated with allogeneic DCs in the presence of pacritinib or DMSO for 5 days, and then cultured with self T-cell responders and fresh DCs without additional drug exposure. The suppressive potency of either pacritinib- or DMSO-treated iTregs was similar, suggesting JAK2 is not required for iTreg function. We then evaluated whether JAK2 inhibition could prevent human skin graft rejection in an NSG mouse xenograft model. A 1x1 cm split-thickness human skin graft was transplanted onto the animal dorsally. The mice rested for 30 days after surgery to permit engraftment, then received 5x106 allogeneic peripheral blood mononuclear cells (PBMC) by intraperitoneal injection. Pacritinib was administered at the same dose and schedule for 2 weeks beginning at time of PBMC injection as tolerated by the NSG mice. The treatment significantly delayed allograft rejection by the human donor PBMCs, compared with vehicle control (Figure 2: Median graft survival 32.5 v 51 days, Day +60 survival 0% v 33.3%, pooled data from 2 experiments, n=5-6 mice per arm, P =.0011). Collectively, these data clearly identify JAK2 as a therapeutic target to control donor alloreactivity and promote iTreg responses after HCT or solid organ transplantation. *BCB and DB contributed equally to this work. Figure 1. Decreased murine GVHD with JAK2 deficient T-cells Figure 1. Decreased murine GVHD with JAK2 deficient T-cells Figure 2. Decreased human skin graft rejection with JAK2 inhibitor, pacritinib Figure 2. Decreased human skin graft rejection with JAK2 inhibitor, pacritinib Disclosures Singer: CTI BioPharma, Corp: Employment, Equity Ownership.


1981 ◽  
Vol 153 (5) ◽  
pp. 1044-1057 ◽  
Author(s):  
B E Loveland ◽  
P M Hogarth ◽  
R Ceredig ◽  
I F McKenzie

The Ly phenotype of cells mediating skin graft rejection was determined using monoclonal anti-Lyt-1.1 and Lyt-2.1 antibodies in CBA mice that received CBA lymphoid cells from mice sensitized to C57BL/6; i.e., alloantigenic differences arising from the H-2 and non-H-2 loci. It was clear that graft rejection was due wholly to the presence of Lyt-1 cells in the inoculum and that Lyt-123 or Lyt-23 cells had no effect. Furthermore, no synergism was noted between Lyt-1 and Lyt-2 cells. In this model, both the cytotoxic T cell and cytotoxic lymphocyte precursors were shown to be Lyt-123 and these could be depleted from sensitized Lyt-1 populations that mediated graft rejection. Thus cytotoxic T cells are not responsible for skin graft rejection, but rather, this is mediated by an Lyt-1 cell. Whether this T cell is distinct from other Lyt-1 cells (T helper, T cells mediating delayed hypersensitivity) is not clear at present, but other evidence, and traditional concepts, link graft rejection and delayed type hypersensitivity as being different manifestations of the same mechanism.


Author(s):  
Suthida Boonsom ◽  
Suda Vannaprasaht ◽  
Wichittra Tassaneeyakul ◽  
Surasakdi Wongratanacheewin ◽  
Cholatip Pongskul ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 232-232
Author(s):  
Ping Zhang ◽  
Jieying Wu ◽  
Divino Deoliveria ◽  
Nelson J. Chao ◽  
Benny J. Chen

Abstract Abstract 232 Several different groups have independently demonstrated that non-allospecific effector memory T cells do not induce graft-versus-host disease (GVHD). Limited data are available regarding the ability of allospecific effector memory T cells to induce GVHD. We first studied this question in the C57BL/6 into BALB/c model. Similar to the data previously published by other groups, purified CD62L- effector memory T cells isolated from donors, who were primed with the host antigens 8 weeks earlier, had decreased ability to induce acute GVHD compared with unseparated and CD62L+ T cells. Similar results were observed when the parous female mice, who were sensitized to the host antigens during pregnancy, were used as memory T cell donors. In order to study this question more definitely and to understand the mechanisms underlying these surprising observations, we further studied the ability of allospecific effector memory T cells to induce GVHD using a novel GVHD model mediated by transgenic TEa T cells. All TEa T cells are CD4+ and recognize the same peptide in the context of I-Ab. This peptide corresponds to positions 52-68 from the alpha-chain of I-E class II molecules and is expressed in all antigen presenting cells from H-2b/I-E+ strains such as CB6F1 mice. To generate memory T cells, naïve TEa cells were first parked in Rag1−/− mice and then immunized with irradiated CB6F1 spleen cells. More than eight weeks later, ∼98% of TEa cells obtained a memory phenotype (CD44high, CD45RB-.CD127+,CD11a bright, FasL bright, Ki67-, CD28-, KLRG-). Of them, about 93% were CD62L-CD44high effector memory T cells and 7% were CD62L+CD44high central memory T cells. These Rag1−/− mice that contained memory phenotype TEa cells rejected CB6F1 skin grafts much faster than naïve TEa mice did (median survival time: 6.5 vs. 13 days, P=0.01), suggesting that the memory phenotype T cells contained in these mice are functional. Moreover, CD62L-CD44high TEa cells purified from these mice mediated faster and stronger in vitro proliferative responses against alloantigens than naïve TEa cells did, further demonstrating that they are true functional effector memory T cells. We next tested the ability of these effector memory TEa cells to induce GVHD. Effector memory TEa cells were obtained after depletion of CD62L+ cells using magnetic beads and the purity was more than 99%. Transplantation of 1′105 TEa naïve T cells together with 1′107 T cell depleted bone marrow cells into lethally irradiated CB6F1 recipients induced lethal GVHD in all recipients and all animals in this group died within 35 days after transplantation. In contrast, none of the effector memory TEa cell recipients developed GVHD and all of them survived more than 100 days post transplantation (P<0.01, compared with naïve T cell control). To understand the mechanisms underlying these observations, we studied the kinetics of TEa proliferative responses upon challenge with alloantigens. The data indicated that effector memory TEa cells reached the peak responses faster than naïve TEa cells did. CFSE tracking experiment further confirmed this observation. Simultaneous staining with anti-Anexin V antibody and 7-AAD demonstrated that effector memory TEa cells undergone apoptosis and died faster than naïve T cells did. In conclusion, these data underscore the fundamental difference of alloresponses mediated by antigen-specific effector memory T cells in graft rejection and GVHD settings. The TEa transgenic T cell skin graft and GVHD models would allow further understanding of the unique alloresponses mediated by allospecific memory T lymphocytes in GVHD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 109 (2) ◽  
pp. 827-835 ◽  
Author(s):  
Dela Golshayan ◽  
Shuiping Jiang ◽  
Julia Tsang ◽  
Marina I. Garin ◽  
Christian Mottet ◽  
...  

Abstract CD4+CD25+ regulatory T (Treg) cells play a critical role in the induction and maintenance of peripheral immune tolerance. In experimental transplantation models in which tolerance was induced, donor-specific Treg cells could be identified that were capable of transferring the tolerant state to naive animals. Furthermore, these cells appeared to have indirect allospecificity for donor antigens. Here we show that in vivo alloresponses can be regulated by donor alloantigen-specific Treg cells selected and expanded in vitro. Using autologous dendritic cells pulsed with an allopeptide from H2-Kb, we generated and expanded T-cell lines from purified Treg cells of CBA mice (H2k). Compared with fresh Treg cells, the cell lines maintained their characteristic phenotype, suppressive function, and homing capacities in vivo. When cotransferred with naive CD4+CD25− effector T cells after thymectomy and T-cell depletion in CBA mice that received CBK (H2k+Kb) skin grafts, the expanded Treg cells preferentially accumulated in the graft-draining lymph nodes and within the graft while preventing CBK but not third-party B10.A (H2k+Dd) skin graft rejection. In wild-type CBA, these donor-specific Treg cells significantly delayed CBK skin graft rejection without any other immunosuppression. Taken together, these data suggest that in vitro–generated tailored Treg cells could be considered a therapeutic tool to promote donor-specific transplant tolerance.


Sign in / Sign up

Export Citation Format

Share Document