TRAIL/ DR5 Interactions Are Important for Thymic Damage After Allogeneic Bone Marrow Transplantation.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 234-234
Author(s):  
Il-Kang Na ◽  
Sydney X Lu ◽  
Nury L. Yim ◽  
Gabrielle L. Goldberg ◽  
Jennifer Tsai ◽  
...  

Abstract Abstract 234 Thymic GVHD (tGVHD) after allogeneic bone marrow transplantation (allo-BMT) is associated with prolonged immunodeficiency. We have previously shown that thymic output after allo-BMT is directly related to thymus size, and inversely related to donor T cell dose and GVHD severity. Additionally, radiation-containing preparative regimens upregulate the death receptors Fas and DR5 on thymic stroma (especially epithelium) while decreasing expression of the anti-apoptotic protein cFLIP, thereby sensitizing the thymus to GVHD. Moreover, small numbers of donor alloreactive T cells are sufficient to cause tGVHD, and they utilize the Fas/Fas ligand (FasL) and TRAIL/DR5 pathways to mediate damage thymic stroma, architecture and function. We performed experiments in both MHC-mismatched and MHC-matched minor antigen-disparate model systems, and demonstrated the exquisite sensitivity of the thymus to as few as 1–2.5×105 donor T cells, which mediated tGVHD without evidence of overt clinical disease or significant weight loss. Additionally, tGVHD is partially reversible in our model systems (contingent on a low T cell dose), such that mice with tGVHD exhibit a transient but partially reversible decrease in thymic cellularity when measured at days 28 vs. 60 post-transplant (Figure 1). To further study the role of TRAIL in tGVHD, we asked whether (1) alloreactive T cells and (2) the inflammation associated with conditioning and acute GVHD, were strictly required for TRAIL/DR5-mediated thymic damage. We treated recipients of T cell-depleted allo-BMT with the amDR5-1 agonistic antibody (0.2 mg i.p. per dose) either in the ‘early‘ peri-transplant period, or ‘late,‘ in the second week post-transplant. Allo-BMT recipients treated ‘early‘ with amDR5-1 had significantly decreased thymic cellularity and splenic BM-derived T cells as compared with controls. Furthermore, we observed similar BM cellularity and BM-derived lineage− sca-1+ckit+ (LSK), all in the absence of donor alloreactive T cells and GVHD (Figure 2). We observed similar results with mice treated ‘late‘ amDR5-1 using the later schedule, which indicates that the thymus has continued sensitivity to TRAIL throughout the post-transplant period, and that GVHD and/or conditioning-associated cytokines are not required to enable TRAIL-mediated damage to the thymus. We further assessed the expression of DR5 on donor BM-derived thymocytes to determine whether amDR5-1 acted directly on thymocytes. We observed that on day 28 after T cell-depleted allo-BMT, only 1-2% of donor thymocytes expressed DR5, suggesting that amDR5-1 (and potentially TRAIL) mediate their effects on thymic cellularity and function primarily via an indirect mechanism (Figure 3). These data suggest to us that significant damage to the thymus and thymopoiesis during allo-BMT: Together, our data in clinically-relevant mouse allo-BMT models suggests that the thymus is highly sensitive to GVHD and endures severe damage at relatively low levels of systemic GVHD. Moreover, post-transplant thymic atrophy is a partially-reversible process which depends on the T cell dose, and which occurs via the TRAIL pathway. Finally, we provide significant mechanistic insight which shows that TRAIL-mediated thymic damage can occur (1) throughout the early post-transplant period, and (2) does not strictly require alloreactive T cells, or the inflammatory processes associated with conditioning and GVHD. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 115 (21) ◽  
pp. 4293-4301 ◽  
Author(s):  
Janelle A. Olson ◽  
Dennis B. Leveson-Gower ◽  
Saar Gill ◽  
Jeanette Baker ◽  
Andreas Beilhack ◽  
...  

Abstract Natural killer (NK) cells suppress graft-versus-host disease (GVHD) without causing GVHD themselves. Our previous studies demonstrated that allogeneic T cells and NK cells traffic similarly after allogeneic bone marrow transplantation (BMT). We therefore investigated the impact of donor NK cells on donor alloreactive T cells in GVHD induction. Animals receiving donor NK and T cells showed improved survival and decreased GVHD score compared with controls receiving donor T cells alone. Donor T cells exhibited less proliferation, lower CD25 expression, and decreased interferon-γ (IFN-γ) production in the presence of NK cells. In vivo, we observed perforin- and Fas ligand (FasL)–mediated reduction of donor T cell proliferation and increased T cell apoptosis in the presence of NK cells. Further, activated NK cells mediated direct lysis of reisolated GVHD-inducing T cells in vitro. The graft-versus-tumor (GVT) effect was retained in the presence of donor NK cells. We demonstrate a novel mechanism of NK cell–mediated GVHD reduction whereby donor NK cells inhibit and lyse autologous donor T cells activated during the initiation of GVHD.


Blood ◽  
2009 ◽  
Vol 113 (7) ◽  
pp. 1574-1580 ◽  
Author(s):  
Robert R. Jenq ◽  
Christopher G. King ◽  
Christine Volk ◽  
David Suh ◽  
Odette M. Smith ◽  
...  

Abstract Keratinocyte growth factor (KGF), which is given exogenously to allogeneic bone marrow transplantation (allo-BMT) recipients, supports thymic epithelial cells and increases thymic output of naive T cells. Here, we demonstrate that this improved T-cell reconstitution leads to enhanced responses to DNA plasmid tumor vaccination. Tumor-bearing mice treated with KGF and DNA vaccination have improved long-term survival and decreased tumor burden after allo-BMT. When assayed before vaccination, KGF-treated allo-BMT recipients have increased numbers of peripheral T cells, including CD8+ T cells with vaccine-recognition potential. In response to vaccination, KGF-treated allo-BMT recipients, compared with control subjects, generate increased numbers of tumor-specific CD8+ cells, as well as increased numbers of CD8+ cells producing interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). We also found unanticipated benefits to antitumor immunity with the administration of KGF. KGF-treated allo-BMT recipients have an improved ratio of T effector cells to regulatory T cells, a larger fraction of effector cells that display a central memory phenotype, and effector cells that are derived from a broader T-cell–receptor repertoire. In conclusion, our data suggest that KGF can function as a potent vaccine adjuvant after allo-BMT through its effects on posttransplantation T-cell reconstitution.


Blood ◽  
1988 ◽  
Vol 71 (5) ◽  
pp. 1196-1200 ◽  
Author(s):  
A Velardi ◽  
A Terenzi ◽  
S Cucciaioni ◽  
R Millo ◽  
CE Grossi ◽  
...  

Abstract Peripheral blood T cell subsets were evaluated in 11 patients during the reconstitution phase after allogeneic bone marrow transplantation and compared with 11 age-matched controls. The proportion of cells coexpressing Leu7 and CD11b (C3bi receptor) markers was determined within the CD4+ (T-helper) and the CD8+ (T-suppressor) subsets by two- color immunofluorescence analysis. CD4+ and CD8+ T cells reached normal or near-normal values within the first year posttransplant. In contrast to normal controls, however, most of the cells in both subsets coexpressed the Leu7 and CD11b markers. T cells with such phenotype display the morphological features of granular lymphocytes (GLs) and a functional inability to produce interleukin 2 (IL 2). These T cell imbalances were not related to graft v host disease (GvHD) or to clinically detectable virus infections and may account for some defects of cellular and humoral immunity that occur after bone marrow transplantation./


Blood ◽  
1996 ◽  
Vol 87 (7) ◽  
pp. 3019-3026 ◽  
Author(s):  
K Kubo ◽  
K Yamanaka ◽  
H Kiyoi ◽  
H Fukutani ◽  
M Ito ◽  
...  

From the viewpoint of T-cell receptor (TCR) repertoire, we studied the role of T cells in acute graft-versus-host disease (GVHD) after allogeneic bone marrow transplantation (allo-BMT) from an HLA-identical sibling. By means of inverse polymerase chain reaction method and DNA sequencing, we analyzed TCR-alpha and -beta transcripts from GVHD lesions and peripheral blood (PB) in a patient with typical GVHD together with PB from donor. At the initial onset of GVHD, V alpha-7 and -19 subfamilies were oligoclonally expanded in the PB compared with those in the oral mucosal lesions. At the second onset, V alpha-2, and V beta-6 subfamilies were more frequently detected in the cutaneous lesion than in the PB. Some TCR transcripts were recurrently found either in the mucosal or cutaneous lesions (or in both) and not in the PB. Furthermore, some of recurrent TCR transcripts in the lesions shared V gene segments and common motifs of complementarity determining region-3. These findings suggested that T cells infiltrating the GVHD lesions recognized a limited kind of antigens presented by patient's tissues with GVHD, and that T-cell repertoire in the GVHD lesions was different from that in the PB.


2020 ◽  
Vol 21 (9) ◽  
pp. 3347
Author(s):  
Jose-Ignacio Rodriguez-Barbosa ◽  
Pascal Schneider ◽  
Luis Graca ◽  
Leo Bühler ◽  
Jose-Antonio Perez-Simon ◽  
...  

Regulatory T cells (Tregs) are essential for the maintenance of tolerance to self and non-self through cell-intrinsic and cell-extrinsic mechanisms. Peripheral Tregs survival and clonal expansion largely depend on IL-2 and access to co-stimulatory signals such as CD28. Engagement of tumor necrosis factor receptor (TNFR) superfamily members, in particular TNFR2 and DR3, contribute to promote peripheral Tregs expansion and sustain their survival. This property can be leveraged to enhance tolerance to allogeneic transplants by tipping the balance of Tregs over conventional T cells during the course of immune reconstitution. This is of particular interest in peri-transplant tolerance induction protocols in which T cell depletion is applied to reduce the frequency of alloreactive T cells or in conditioning regimens that allow allogeneic bone marrow transplantation. These conditioning regimens are being implemented to limit long-term side effects of continuous immunosuppression and facilitate the establishment of a state of donor-specific tolerance. Lymphopenia-induced homeostatic proliferation in response to cytoreductive conditioning is a window of opportunity to enhance preferential expansion of Tregs during homeostatic proliferation that can be potentiated by agonist stimulation of TNFR.


Blood ◽  
2006 ◽  
Vol 107 (5) ◽  
pp. 2045-2051 ◽  
Author(s):  
Barry J. Kappel ◽  
Javier Pinilla-Ibarz ◽  
Adam A. Kochman ◽  
Jeffrey M. Eng ◽  
Vanessa M. Hubbard ◽  
...  

Major histocompatibility complex (MHC) molecules carrying selected peptides will bind specifically to their cognate T-cell receptor on individual clones of reactive T cells. Fluorescently labeled, tetrameric MHC-peptide complexes have been widely used to detect and quantitate antigen-specific T-cell populations via flow cytometry. We hypothesized that such MHC-peptide tetramers could also be used to selectively deplete unique reactive T-cell populations, while leaving the remaining T-cell repertoire and immune response intact. In this report, we successfully demonstrate that a tetramer-based depletion of T cells can be achieved in a murine model of allogeneic bone marrow transplantation. Depletion of a specific alloreactive population of donor splenocytes (< 0.5% of CD8+ T cells) prior to transplantation significantly decreased morbidity and mortality from graft-versus-host disease. There was no early regrowth of the antigen-specific T cells in the recipient and in vivo T-cell proliferation was greatly reduced as well. Survival was increased more than 3-fold over controls, yet the inherent antitumor activity of the transplant was retained. This method also provides the proof-of-concept for similar strategies to selectively remove other unwanted T-cell clones, which could result in novel therapies for certain autoimmune disorders, T-cell malignancies, and solid organ graft rejection.


Blood ◽  
2005 ◽  
Vol 106 (9) ◽  
pp. 3300-3307 ◽  
Author(s):  
Christian A. Wysocki ◽  
Qi Jiang ◽  
Angela Panoskaltsis-Mortari ◽  
Patricia A. Taylor ◽  
Karen P. McKinnon ◽  
...  

AbstractCD4+CD25+ regulatory T cells (Tregs) have been shown to inhibit graft-versus-host disease (GVHD) in murine models, and this suppression was mediated by Tregs expressing the lymphoid homing molecule l-selectin. Here, we demonstrate that Tregs lacking expression of the chemokine receptor CCR5 were far less effective in preventing lethality from GVHD. Survival of irradiated recipient animals given transplants supplemented with CCR5-/- Tregs was significantly decreased, and GVHD scores were enhanced compared with animals receiving wild-type (WT) Tregs. CCR5-/- Tregs were functional in suppressing T-cell proliferation in vitro and ex vivo. However, although the accumulation of Tregs within lymphoid tissues during the first week after transplantation was not dependent on CCR5, the lack of function of CCR5-/- Tregs correlated with impaired accumulation of these cells in the liver, lung, spleen, and mesenteric lymph node, more than one week after transplantation. These data are the first to definitively demonstrate a requirement for CCR5 in Treg function, and indicate that in addition to their previously defined role in inhibiting effector T-cell expansion in lymphoid tissues during GVHD, later recruitment of Tregs to both lymphoid tissues and GVHD target organs is important in their ability to prolong survival after allogeneic bone marrow transplantation.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 251-251 ◽  
Author(s):  
Alan Hanash ◽  
Robert B. Levy

Abstract Despite the potential to cure both acquired and inherited disorders involving the hematopoietic compartment, application of allogeneic bone marrow transplantation (BMT) is limited by the frequent and severe outcome of Graft vs. Host Disease (GVHD). Unfortunately, efforts to reduce GVHD by purging the donor graft of T cells have resulted in poor engraftment and elevated disease recurrence. Alternative cell populations capable of supporting allogeneic engraftment without inducing GVHD could increase the potential for donor-recipient matching and decrease treatment associated risks. We have observed that GVHD-suppressive donor CD4+CD25+ T cells are capable of supporting allogeneic hematopoietic engraftment, as demonstrated by initial donor progenitor activity and long-term chimerism and tolerance. Using a murine MHC mismatched model transplanting 0.5–2x106 GFP+ C57BL/6 (B6) T cell-depleted bone marrow cells into 7.0 Gy sublethally irradiated BALB/c recipients, splenic CFU assessment demonstrated that co-transplantation of 1x106 B6 CD4+CD25+ T cells lead to increased donor lineage-committed GM (p&lt;.01) and multi-potential HPP (p&lt;.05) progenitors seven days post-BMT compared to transplantation of BM alone. Furthermore, co-transplantation of CD4+CD25+ T cells lead to lymphoid and myeloid chimerism in peripheral blood (lineage specific mean donor chimerism ± SE: B220, 67.7±15.2 vs. 0.3±0.3; CD4, 38.3±10.5 vs.0.9±0.9; CD8, 48.3±11.0 vs. 1.0±1.0; Mac-1, 58.8±16.5 vs. 0.3±0.3) and the presence of donor GM and HPP progenitors in recipient marrow two months post-BMT (mean CFU chimerism ± SE: CFU-GM, 54.5±12.8 vs. 0.0; CFU-HPP, 63.0±17.8 vs.0.0). Donor chimerism persisted six months post-BMT and was associated with tolerance to donor and host antigens by acceptance of donor and host skin grafts &gt;50 days post-homotopic grafting. Characterization of the initial invents of engraftment support demonstrated that augmentation of donor progenitors did not require CD4+CD25+ T cell IL-10, as co-transplantation of B6-wt and B6-IL-10−/− CD4+CD25+ T cells both significantly increased total CFU-GM (mean CFU±SE: BM alone, 657.5±248.2; BM + wt, 1972±331.5; BM + IL-10−/−, 1965±401.7; both p&lt;.05 vs. BM alone). Assessment of the antigenic requirements for activation of progenitor support demonstrated that donor CD4+CD25+ T cells did not require alloreactivity to support progenitors, as BALB/c x B6 F1 CD4+CD25+ T cells significantly increased B6 CFU-GM in BALB/c recipients (p&lt;.001 vs. BM alone). However, B6 CD4+CD25+ T cells failed to augment C3H/HeJ CFU-GM in BALB/c recipients (p&gt;.05 vs. BM alone), suggesting that donor CD4+CD25+ T cells might require recognition of syngeneic MHC for progenitor support. Indeed, augmentation of donor CFU-GM was abrogated when B6 CD4+CD25+ T cells were co-transplanted with B6-MHC class II−/− marrow into BALB/c recipients (p&gt;.05 vs. BM alone). In conclusion, donor CD4+CD25+ T cells capable of promoting long-term engraftment and tolerance do not require IL-10 for support of initial donor progenitor activity, however progenitor support does require co-transplantation with syngeneic MHC class II expressing marrow. Donor CD4+CD25+ T cells may thus represent a useful alternative to unfractionated T cells for promotion of engraftment following allogeneic hematopoietic transplantation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4610-4610
Author(s):  
Kataryna A. Darlak ◽  
Jian-Ming Li ◽  
Wayne Harris ◽  
Edmund K. Waller

Abstract Background: Allogeneic hematopoietic stem cell (HSC) transplant is a treatment used to cure patients with high risk and relapsed acute leukemia, via a graft versus leukemia (GvL) effect. In our previous studies, depletion of CD11b+ cells (containing CD11b+ dendritic cells, CD11b+ NK cells, and myeloid suppressor progenitor cells) from donor BM improved immune reconstitution and enhanced GvL effects without increased rates of GvHD in mice (Li, et al. BBMT 2004). In order to elucidate which subset of donor CD11b+ cells was responsible for this effect, we have transplanted combinations of FACS purified HSC, donor T cells, and CD11b- dendritic cells (DC) and found similar improvement in GvL activity associated with donor T cells polarized towards a Th1 phenotype (Li, et al. Blood 2007). In contrast, grafts containing FACS purified HSC, donor spleen T cells, and CD11b+ DC did not have significant GvL activity and donor T cells were polarized towards a Th2 phenotype. The objective of this study was to determine if selective depletion of CD11b+ dendritic cells by FACS would be a clinically relevant method, producing similar results as engraftment with purified populations of HSC, Cd11b- DC, and T cells. Methods: Selective CD11b+ DC depletion was achieved by FACS sorting in which cells from BM were sorted following gating of all nucleated cells with the exception of the CD11b+CD11c+Lineage- cells. The CD11b+ cells comprised ~1% of the BM. Undepleted BM was also stained and sorted using only a light scatter gate as a control for stress encountered during sorting. To study the long term effects of specific CD11b+ DC depletion, B10.BR or BA.B10 recipients were transplanted with 5×10^6 CD11b+ DC FACS-depleted or undepleted BM cells and 1×10^6 spleen T cells from C57BL/6J, Balb/C, and BA donors. Recipient mice were irradiated with 11Gy one day prior to the transplant. Recipient mice were monitored for survival, weight change, and GvHD score (based on weight change, activity, posture, fur texture, and skin condition) throughout the duration of the experiment and for chimerism of engraftment at days 30, 60, and 100 post transplant. Donor T cells were recovered from transplant recipients on days 3 and 10 post transplant and were examined for their Th1/Th2 polarization by flow cytometry after intracellular cytokine staining and ELISA of supernatants following short term culture. Results: B10.BR mice receiving undepleted BM from C57BL/6J or Balb/C mice had 100 day survivals of 60% and 40% respectively. Mice receiving CD11b+ DC depleted BM from C57BL/6J or Balb/C mice had equivalent survival at 80% and 60% survival, respectively, compared with mice receiving undepleted BM (p=ns). T cell chimerism at day 100 was over 95% donor chimerism for all mice, regardless of CD11b+ DC depletion. Differences in concentration of donor T cells in the blood were not significant between CD11b+ DC depleted and undepleted groups. The level of GvHD in recipient mice also did not differ significantly between groups. At day 3 post transplant, intracellular cytokine staining of donor T cells from mice receiving CD11b+ DC depleted BM had significantly higher levels of TNF-alpha compared with donor T cells from recipients of undepleted BM (p&lt;0.05). Levels of IL-4, IL-10, IL-5 and IFN-gamma among donor spleen T cell population were not significantly different between undepleted and CD11b+ depleted BM recipients. Conclusions: Selective depletion of CD11b+ DC by FACS may be a clinically relevant method for increasing GvL activity without increasing GvHD. Depletion of CD11b+ DC from BM grafts polarizes donor T cells towards a Th1 phenotype, without increasing GvHD or affecting survival in recipient mice as compared with mice receiving undepleted BM grafts. The GvL effect of selective CD11b+ FACS depletion will be discussed.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4279-4279
Author(s):  
Kate A Markey ◽  
Rachel D Kuns ◽  
Renee J Robb ◽  
Motoko Koyama ◽  
Kate Helen Gartlan ◽  
...  

Abstract Allogeneic bone marrow transplantation (BMT) remains the therapy of choice for many haematological malignancies, but despite the curative benefit of the immunological graft-versus-leukemia (GVL) effect, relapse remains a key cause of death. We have investigated the role of recipient dendritic cells (DC) in antigen presentation to donor CD8 cytotoxic T cells (CTL) in a model of BMT where GVHD and GVL are directed to multiple minor histocompatibility antigens (mHA) and survival reflects GVL activity. C3H.Sw bone marrow and purified CD8 T cell grafts were transplanted with B6-derived MLL-AF9 induced primary acute myeloid leukemia (AML) into lethally irradiated B6.CD11c.DOG recipients (diphtheria toxin receptor (DTR), ovalbumin and GFP expression driven off the CD11c promoter) such that recipient DC can be deleted by DT administration. Surprisingly, depletion of recipient DC resulted in improved leukemic control (median survival 43 vs 31 days, P <0.001). The use of IRF8-/- BMT recipients (in which the CD8+ DC subset is absent) confirmed that recipient CD8+ DC were critical for regulating these GVL effects (median survival 43 vs 34 days, P = 0.0005). Conversely, when recipient CD8+ DC were expanded in a B6 to B6D2F1 model with bcr-abl/Nup98-HoxA9 induced primary AML, by using Flt3-L treatment for 10 days prior to BMT, GVL effects were completely eliminated, rendering relapse rate equivalent to that seen in the recipients of T cell depleted (TCD) grafts (median survival 11 days in BM+T and TCD groups where recipients were pre-treated with Flt3-L, vs. >45 days in the saline treated BM+T group). The use of B6.CD11c-Rac1 transgenic BMT recipients (who cannot process and present exogenously acquired antigen) confirmed that this effect was the result of endogenous alloantigen presentation by recipient DC and independent of cross-presentation.Using the same depletion strategies in an antigen-specific model (with donor OT-I T cells and B6.CD11c.DOG x DBA/2 F1 recipients) we confirmed that recipient DC invoked effector donor CTL activation, differentiation (CD25+ CD69+ CD62L-) and subsequent apoptosis (as measured by Annexin V; 52.4% vs. 23.9% in DC replete vs. depleted recipients, P = 0.01). There was a consequent profound contraction of the donor CTL compartment by day 10 in DC replete recipients. This contraction of the CTL compartment was associated with reduced expression of the cytolytic molecule granzyme B (MFI 1922 vs 1097, P = 0.02). Antigen presentation has a critical role in the initiation of donor T cell alloreactivity and GVL after BMT. Here we demonstrate that endogenous alloantigen presentation by recipient CD8+ DC to donor T cells leads to activation induced death of donor CTL early after BMT, which in turn facilitates leukemic relapse. This concept has critical implications for the design of therapies that target DC in the peri-transplant period and confirms that recipient DC regulate GVL effects. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document