scholarly journals Prevention of Graft-Versus-Host Disease in Mice Using a Suicide Gene Expressed in T Lymphocytes

Blood ◽  
1997 ◽  
Vol 89 (12) ◽  
pp. 4636-4645 ◽  
Author(s):  
José L. Cohen ◽  
Olivier Boyer ◽  
Benoı̂t Salomon ◽  
Rosine Onclercq ◽  
Frédéric Charlotte ◽  
...  

Abstract Alloreactive T cells present in a bone marrow transplant are responsible for graft-versus-host disease (GVHD), but their depletion is associated with impaired engraftment, immunosuppression, and loss of the graft-versus-leukemia effect. We developed a therapeutic strategy against GVHD based on the selective destruction of these alloreactive T cells, while preserving a competent T-cell pool of donor origin. We generated transgenic mice expressing in their T lymphocytes the Herpes simplex type 1 thymidine kinase (TK) suicide gene that allows the destruction of dividing T cells by a ganciclovir treatment. T cells expressing the TK transgene were used to generate GVHD in irradiated bone marrow grafted mice. We show that a short 7-day ganciclovir treatment, initiated at the time of bone marrow transplantation, efficiently prevented GVHD in mice receiving TK-expressing T cells. These mice were healthy and had a normal survival. They maintained a T-cell pool of donor origin that responded normally to in vitro stimulation with mitogens or third party alloantigens, but were tolerant to recipient alloantigens. Our experimental system provides the proof of concept for a therapeutic strategy of GVHD prevention using genetically engineered T cells.

Blood ◽  
2001 ◽  
Vol 97 (8) ◽  
pp. 2506-2513 ◽  
Author(s):  
William R. Drobyski ◽  
Herbert C. Morse ◽  
William H. Burns ◽  
James T. Casper ◽  
Gordon Sandford

Abstract Donor T cells play a pivotal role in facilitating alloengraftment but also cause graft-versus-host disease (GVHD). Ex vivo T-cell depletion (TCD) of donor marrow is the most effective strategy for reducing GVHD but can compromise engraftment. This study examined an approach whereby donor T cells are selectively eliminated in vivo after transplantation using transgenic mice in which a thymidine kinase(TK) suicide gene is targeted to the T cell using a CD3 promoter/enhancer construct. Lethally irradiated B10.BR mice transplanted with major histocompatibility complex (MHC)–incompatible TCD C57BL/6 (B6) bone marrow (BM) plus TK+ T cells were protected from GVHD after treatment with ganciclovir (GCV) in a schedule-dependent fashion. To examine the effect of GCV treatment on alloengraftment, sublethally irradiated AKR mice underwent transplantation with TCD B6 BM plus limiting numbers (5 × 105) of B6 TK+ T cells. Animals treated with GCV had comparable donor engraftment but significantly reduced GVHD when compared with untreated mice. These mice also had a significantly increased number of donor splenic T cells when assessed 4 weeks after bone marrow transplantation. Thus, the administration of GCV did not render recipients T-cell deficient, but rather enhanced lymphocyte recovery. Adoptive transfer of spleen cells from GCV-treated chimeric mice into secondary AKR recipients failed to cause GVHD indicating that donor T cells were tolerant of recipient alloantigens. These studies demonstrate that administration of TKgene–modified donor T cells can be used as an approach to mitigate GVHD without compromising alloengraftment.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2132-2132
Author(s):  
Eva AS Koster ◽  
Liesbeth C. de Wreede ◽  
Sylwia Wallet-Malicka ◽  
Lisette Bogers ◽  
Peter van Balen ◽  
...  

Abstract After allogeneic stem cell transplantation (alloSCT), donor T cells targeting patient derived hematopoietic cells can induce a Graft versus Leukemia (GvL) effect preventing relapse. However, targeting of healthy patient tissues can cause Graft versus Host Disease (GvHD). The inflammatory environment induced by pre transplantation conditioning, the number of donor T cells in the graft, genetic disparity between patient and donor and the presentation of allo-antigens by activated patient derived antigen-presenting cells (APC) to donor T cells play a role in the development of GvL and/or GvHD. Donor T cell depletion (TCD) reduces GvHD and GvL. After TCD alloSCT, postponed prophylactic donor lymphocyte infusions (pDLI) are often needed to induce a GvL effect. When using 10/10 matched donors, our first dose of pDLI at six months after TCD alloSCT contained 3.0x10^6 T cells/kg (related donor, RD) or 1.5x10^6 T cells/kg (unrelated donor, UD). We evaluated whether the risk of developing GvHD after DLI is influenced by the donor type, intensity of the conditioning and/or patient bone marrow (BM) chimerism at time of DLI Sixty patients with acute leukemia (52 AML, 8 ALL; median age 57; 27 RD, 33 UD) received pDLI at a median of 6.4 months after TCD alloSCT in the absence of GVHD or relapse. Twenty-four patients received myeloablative (MA) conditioning consisting of cyclophosphamide and TBI. 36 patients received non-myeloablative (NMA) conditioning based on fludarabin and busulphan. TCD was performed by adding 20mg alemtuzumab to the graft. Only MA conditioned patients with an UD (n=12) received post transplantation ciclosporin as GvHD prophylaxis, which was tapered from 1 month after alloSCT. Clinically significant GvHD was defined as need of therapeutic systemic immunosuppression (tIS) for GvHD for at least 2 weeks or until death. Bone Marrow (BM) chimerism was measured prior to DLI. Three categories of patient chimerism levels were defined: no patient derived cells (absent), patient derived cells present, but < 5% (low), or ≥ 5% (high). In case of persisting or increasing patient chimerim after pDLI, a second DLI was given at 3-6 months after the first. A multi-state model was designed (Figure 1) with the first DLI (DLI1) as starting state and time. Patients starting tIS after DLI1 transit to the state tIS. Patients who need a second DLI, develop a relapse or die, transit to these respective states. Patients who stay in the state of DLI1 are considered to have a positive outcome. All patients had a follow-up of at least one year after DLI. Numbers in the boxes in Figure 1 represent the number of patients in that state at 1 year after DLI1 and numbers next to the arrows indicate the numbers of patients who made the transition between the two states. Donor type (unrelated versus related), conditioning (NMA versus MA) and patient BM chimerism at time of DLI were included in a Cox model for the transition hazards to investigate their association with the development of GvHD after DLI. For the total group, the cumulative incidence of tIS at 1 year after pDLI was 33% (95% CI 21-45%). Patients with an UD had a hazard ratio (HR) of 1.1 (95% CI 0.4-3.3) of needing tIS after DLI1 compared to patients with a RD. Compared to MA conditioning, NMA conditioned patients had a hazard ratio of 2.1 (95% CI 0.5-8.9) of needing tIS after DLI. They had a HR of 0.2 (95% CI 0.04-0.95) of stopping tIS compared to MA conditioned patients, indicating that DLI after NMA conditioning is associated with more severe GVHD. We hypothesized that this was due to the persistence of patient derived APC. BM chimerism at time of DLI was measured in 47 patients. After NMA and MA conditioning, BM patient chimerism was absent in 14% and 56%, low in 41% and 39%, and high in 45% and 6%, respectively (Fisher's exact test p=0.002 for difference between type of conditioning). Compared to the group without patient chimerism, the low and high patient chimerism group had a HR of 1.9 (95% CI 0.9-4.2) and 3.6 (95% CI 1.7-8.0) of needing tIS after DLI, respectively (Figure 2), demonstrating that the level of patient chimerism is a strong predictor for development of GvHD after DLI, even when taking into account the type of conditioning regimen. Patient BM chimerism at time of pDLI is a strong and independent predictor for the risk of developing GvHD. Dose reduction in case of an UD equalized the GvHD risk compared to a RD. When choosing a T cell dose for pDLI, patient chimerism should be considered a relevant parameter. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1991 ◽  
Vol 77 (11) ◽  
pp. 2515-2523
Author(s):  
RL Truitt ◽  
AA Atasoylu

Graft rejection, mixed chimerism, graft-versus-host disease (GVHD), leukemia relapse, and tolerance are interrelated manifestations of immunologic reactivity between donor and host cells that significantly affect survival after allogeneic bone marrow transplantation (BMT). In this report, a mouse model of BMT, in which the donor and host were compatible at the major histocompatibility complex (MHC), was used (1) to examine the interrelationship of pretransplant conditioning and T- cell content of donor BM with regard to lymphoid chimerism and GVHD and (2) to determine how these factors affected graft-versus-leukemia (GVL) reactivity and donor-host-tolerance. AKR (H-2k) host mice were administered optimal or suboptimal total body irradiation (TBI) as pretransplant conditioning followed by administration of BM cells from B10.BR (H-2k) donor mice with or without added spleen cells as a source of T lymphocytes. Transplanted mice were injected with a supralethal dose of AKR leukemia cells 20 and 45 days post-BMT to assess GVL reactivity in vivo. The pretransplant conditioning of the host and T- cell content of the donor marrow affected the extent of donor T-cell chimerism and the severity of GVH disease. GVL reactivity was dependent on transplantation of mature donor T cells and occurred only in complete chimeras. Transplantation of T-cell-deficient BM resulted in the persistence of host T cells, ie, incomplete donor T-cell chimerism, even when lethal TBI was used. Mixed chimerism was associated with a lack of GVL reactivity, despite the fact that similar numbers of donor T cells were present in the spleens of mixed and complete chimeras. In this model, moderate numbers of donor T cells facilitated complete donor T-cell engraftment, caused only mild GVHD, and provided a significant GVL effect without preventing the subsequent development of tolerance after conditioning with suboptimal TBI. In contrast, severe, often lethal, GVHD developed when the dose of TBI was increased, whereas tolerance and no GVH/GVL reactivity developed when the T-cell content of the marrow was decreased.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1156-1156
Author(s):  
Amritha Balakrishnan ◽  
Burhan Jama ◽  
Nicholas Joseph Gloude ◽  
Eric Jon Anderson ◽  
Edward D. Ball ◽  
...  

Abstract Evidence from clinical investigations and animal models indicate that chronic graft versus host disease (cGVHD) results from defective thymic generation of functional and self-tolerant T cell populations following hematopoietic stem cell transplantation (HSCT). We have previously demonstrated that the rare subset of T cells that naturally express 2 T cell receptors (TCRs) on the cell surface as a result of incomplete allelic exclusion are predisposed to respond to auto- and alloantigens. Dual TCR T cells disproportionately participate in pathologic alloreactivity in HSCT patients and mouse models of acute GVHD. These findings, combined with observations demonstrating that dual TCR T cells represent a physiologic reservoir of unique TCRs that evade negative selection, prompted us to examine the role of thymic selection and dual TCR T cells in cGVHD. To study the role of post-transplant thymopoiesis in generation of potentially pathogenic dual TCR T cells, we used a mouse model of syngeneic bone marrow transplantation into lethally-irradiated recipients. Radiation-induced damage to the thymic stroma was characterized by disruption of thymic architecture and loss of cortical and medullary thymic epithelial cells (TECs). This damage resulted in significantly increased generation of dual TCR T cells following transplantation of congenically-marked syngeneic T cell-depleted bone marrow. Two-fold increased production of dual TCR T cells persisted for at least 20 weeks after transplantation. These data demonstrate the hazard for production of T cells predisposed to pathogenic reactivity in the post-transplant environment, and suggest that dual TCR T cells could be a source of T cells causing cGVHD. To examine involvement of dual TCR T cells in cGVHD, we analyzed peripheral blood samples from patients after allogeneic HSCT (> 12 months post-transplant) using our previously utilized pair-wise TCRVa labeling flow cytometry approach. Flow cytometry analysis revealed that dual TCR T cells were present at increased frequencies in patients with cGVHD (n = 10, 8.3% + 1.1%, P = 0.028) compared to patients without cGVHD (n = 3, 2.5 + 1.1%) or healthy age-matched controls (n = 5, 1.9 + 0.4%). Dual TCR T cells from patients with cGVHD had an activated CD69+ phenotype as compared to T cells expressing only a single TCR from the same patient. Single-cell TCRa/TCRb sequencing confirmed the increased frequencies of dual TCR T cells specific to activated T cells in patients with cGVHD. Repertoire analysis of TCRs sequenced from single cells indicated that the increase in dual TCR T cells was polyclonal. The single-cell sequencing approach enabled multiplexed examination of T cell lineage-associated transcription factors and cytokines. Single-cell transcriptional profiling demonstrated that dual TCR T cells demonstrated predominantly pro-inflammatory and cytotoxic phenotypes with expression of Tbet and perforin. This is in contrast to T cells expressing only a single TCR from the same patient, or dual TCR T cells from healthy control patients, which had a quiescent phenotype. These data indicate a role for dual TCR T cells in mediating cGVHD. Together, these results suggest that dual TCR T cells may be an important link between post-transplant T cell development and cGVHD. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 115 (16) ◽  
pp. 3390-3397 ◽  
Author(s):  
Laurent Burnier ◽  
François Saller ◽  
Linda Kadi ◽  
Anne C. Brisset ◽  
Rocco Sugamele ◽  
...  

Abstract Growth arrest-specific gene 6 (Gas6) is expressed in antigen-presenting cells and endothelial cells (ECs) but not in T cells. When wild-type (WT) or Gas6−/− mice received allogeneic non–T cell–depleted bone marrow cells, hepatic graft-versus-host disease (GVHD) was alleviated in Gas6−/− recipients regardless of donor genotype, but not in WT recipients. T-cell infiltration was more prominent and diffuse in WT than in Gas6−/− recipients' liver. When mice received 0.5 × 106 allogeneic T cells with T cell–depleted allogeneic bone marrow, clinical signs indicated that GVHD was less severe in Gas6−/− than in WT recipients, as shown by a significant improvement of the survival and reduced liver GVHD. These data demonstrate that donor cells were not involved in the protection mechanism. In addition, lack of Gas6 in antigen-presenting cells did not affect WT or Gas6−/− T-cell proliferation. We therefore assessed the response of WT or Gas6−/− ECs to tumor necrosis factor-α. Lymphocyte transmigration was less extensive through Gas6−/− than WT ECs and was not accompanied by increases in adhesion molecule levels. Thus, the lack of Gas6 in ECs impaired donor T-cell transmigration into the liver, providing a rationale for considering Gas6 pathway as a potential nonimmunosuppressive target to minimize GVHD in patients receiving allogeneic hematopoietic stem cell transplantation.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1749-1749
Author(s):  
Bondanza Attilio ◽  
Valtolina Veronica ◽  
Magnani Zulma ◽  
La Seta Catamancio Simona ◽  
Benati Claudia ◽  
...  

Abstract Background. Suicide gene therapy is a promising approach for the safe exploitation of the graft-versus-leukemia effect. The insertion of Herpes Simplex Virus thymidine kinase confers an inducible suicidal phenotype upon ganciclovir (GCV) administration, thus enabling the selective elimination of T lymphocytes causing graft-versus-host disease (GvHD). Despite clinical and experimental studies substantiating the efficacy of the strategy, protocols to generate genetically modified cells (GMC) has been shown to reduce alloreactivity. The physiological CD4/CD8 ratio is inverted and GMC are enriched for “effector memory” T cells. Co-stimulation through CD28 has been shown to preserve the functional phenotype GMC. XcyteTM Dynabeads®, 4,5 μm anti-CD3 and anti-CD28 coated paramagnetic beads (bCD3/CD28) sustain T cell proliferation and can be used to obtain GMC. Aim. To in vitro characterize human suicide GMC generated with bCD3/CD28 GMC (XcyteTM Dynabeads®, Xcyte Therapies, Inc.) and to test their ability to engraft and cause GvHD in a xenogeneic mouse model. Results. bCD3/CD28 (bead to T cell ratio 3:1) are a potent stimulus for cell cycle entry for both CD4+ and CD8+ human T cells. This permits retroviral transduction (SFCMM#3 vector, Molmed SpA) and preservation of CD4/CD8 ratio. GMC generated with bCD3/CD28 are enriched for “central memory” T cells (CD45RA+CCR7+ 34±7%, CD28+CD27+ 67±12%, intracytoplasmic IL-2+ 14±5%, IFN-γ+ 10±3% and perforin+ 7±3%) when compared with GMC generated with anti-CD3 (CD3) alone (CD45RA+CCR7+ 17±4%, CD28+CD27+ 21±5%, intracytoplasmic IL-2+ 5±3%, IFN-γ+ 52±11% and perforin+ 22±4%). bCD3/CD28 GMC resist activation induced cell death (AxV+PI+ 12±3% vs 42±13% for CD3 GMC). When injected i.p. in NOD/SCID mice conditioned with irradiation and anti-NK depleting antibodies bCD3/CD28 GMC engraft with a faster kinetics (human chimerism at 2 weeks 14±7%) than observed for for CD3 GMC (5±2%). In this model, mice injected with unmodified human lymphocytes develop signs of xenogeneic (X-) GvHD (ruffled fur, hunched back, weight loss and finally death with massive accumulation of human T cell in lymphoid organs) by week 5. X-GvHD observed in mice injected with CD3 GMC has a significant slower course with a proportion of mice surviving week 8. X-GvHD caused by bCD3/CD28 GMC kill all the animals by week 7 (p<0,05 vs CD3 GMC). In mice with established X-GvHD caused by GMC treatment with GCV leads to a reduction in circulating GMC and modulates X-GvHD. GCV administration is not able to cure animals suffering from X-GvHD caused by unmodified T lymphocytes. Conclusions. GMC generated with bCD3/CD28 display a “central memory” functional phenotype and are significantly more efficient than CD3 GMC in causing lethal X-GvHD. GCV administration is able to abrogate X-GvHD caused by GMC. These results validate a tool for the generation of human suicide GMC with high alloreactive potential to be utilized in clinical protocols of adoptive immunotherapy of tumors.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3740-3740
Author(s):  
Bindu Kanathezhath ◽  
Sandra K Larkin ◽  
Julika Kaplan ◽  
Mark C. Walters ◽  
Frans Kuypers

Abstract Abstract 3740 Graft-versus-Host disease (GVHD) causes significant morbidity and mortality after allogeneic transplantation. Regulatory T cells of both donor and host origin can limit GVHD. Acute GVHD early post transplantation is heralded by a cytokine storm induced by a dominant T helper type 1(Th1) response, which damages host tissues like skin, gut, liver and lungs. We hypothesized that co-transplantation of photochemically (S-59) treated T cells modulates T cell effecter subsets and their cytokine milieu, and thereby reduces acute GVHD. To this end, we transplanted whole bone marrow cells in a major histocompatibility complex (MHC) antigen mismatched murine model using marrow with T cells from C57BL/6J AKR (acute GVHD model), with and without S-59 treatment. We observed equivalent elevation of CD4+Th1 (IL-2 and IFN-g), cytotoxic CD8+ (FAS, IFN-g and TNF-a) and CD4+Th2 (IL-4, IL-5, IL-6, IL-13) cytokines in recipients of both groups during the first week after transplantation. While the Th1 cytokines persisted as long as 10 days after transplantation, there was a shift to a regulatory T cell (Treg) cytokine profile (Transforming growth factor b [TGF-b] and IL-10) in the group that received the S-59 treated T cells. TGF-b and IL-10 levels were higher in the peripheral blood and bone marrow of the study group compared to controls (table). This was accompanied by the appearance of FoxP3High CD4+ CD25+ Tregs in the spleen and CD4+ Th17 cytokine (IL-17) elevation in the thymic compartment of recipients that received S-59 treated T cells (mean-28.44pg/ml versus 1.45pg/ml, p=0.0059). In-vivo tracking of S-59 treated T cells demonstrated the disappearance of these cells in the peripheral blood, spleen, bone marrow and thymus by 48 hours after transplantation. Nonetheless, we noted that recipients of S-59 treated T cells had significantly less acute GVHD and better overall survival (p=0.0001). In summary, our experiments indicate that there is an initial dominance of inflammatory and CD4 Th1 cytokines immediately post transplantation. Co-transplantation of S-59 treated T cells shifts the effecter CD4 T cell profile to resemble a Treg phenotype. Despite the absence of circulating photochemically treated T cells, significant alterations in the recipient cytokine milieu persisted after transplantation. Thus, S-59 treated T cells appear to exert an important immunomodulatory effect to ameliorate GVHD and improve survival after MHC-mismatched allogeneic transplantation. Blood (pg/ml) Bone marrow (pg/ml) Thymus (pg/ml) TGF-b [S59] 8482.7 * 7062.1 2926.9 TGF-b [control] 1.1 301.55 46.5 IL-10 [S59] 419.0 ** 174.0 30.4 IL-10 [control] <1 82.1 33.44 ρ value- * 0.0001 ** 0.0006 Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4479-4479
Author(s):  
Kathryn W Juchem ◽  
Britt Anderson ◽  
Cuiling Zhang ◽  
Arlene Sharpe ◽  
Jennifer McNiff ◽  
...  

Graft-versus-host disease (GVHD) is a complication of allogeneic stem cell transplantation (alloSCT). In murine models of alloSCT, naive T cells (TN) cause GVHD while effector memory T cells (TEM) do not. To determine why TEM fail to cause GVHD, we generated a novel T-cell receptor transgenic GVHD model. In this model CD4+ TS1 T cells, which recognize an epitope of influenza hemagglutinin (HA), are transferred, along with syngeneic bone marrow, into irradiated transgenic recipients that express HA in all tissues (HA104 Tg mice). We found that TS1 TN induced early and prolonged weight loss and caused GVHD-like pathology in the skin, liver and colon. In contrast, TS1 TEM induced mild, transient weight loss and minimal pathology, demonstrating that TEM have repertoire-independent characteristics that limit their ability to induce GVHD. Post transplant analysis revealed that TS1 TEM progeny, relative to TS1 TN progeny, produced less IFN-γ, proliferated and accumulated less in the colon, and expressed higher levels of the inhibitory molecule PD-1. To investigate whether PD-1 was responsible for limiting pathogenesis by TEM, we used hosts and donor bone marrow lacking both PD-L1 and PD-L2. The absence of PD-L1/2 did not enable TS1 TEM to cause early weight loss. However, between 35 and 60 days post transplant, TS1 TEM recipients lacking PD-L1/2 rapidly began losing weight and approximately 50% died. Weight loss in TEM recipients was dependent upon lack of PD-L1/2 expression on both donor bone marrow and host cells, including radioresistant stromal cells, suggesting a possible role for PD-L1/2 expressed in tissues. Indeed, global absence of PD-L1 alone, which (in contrast to PD-L2) is expressed on parenchymal tissues, also resulted in late weight loss in recipients given TEM. To determine the reason for late weight loss, we surveyed tissue histopathology. Surprisingly, in the absence of PD-L1/2, TEM recipients did not develop exacerbated colon pathology but instead developed mononuclear infiltrates and mycocyte necrosis in the heart, accompanied by heart block and decreased cardiac output. Interestingly, heart disease was also seen in PD-L1/2 deficient TN recipients that survived to later time points, indicating that the protective role of PD-L1/2 applied more generally to GVHD induced by CD4 T cells. Strikingly, the extensive infiltrates in affected hearts were mostly comprised of non-TS1 T cells, including both CD4 and CD8 cells. These cells are likely host-derived, as severe cardiac infiltrates were seen when Rag-deficient donor BM was used to reconstitute host hematopoiesis. We therefore hypothesize that in GVHD PD-L1/2 normally prevent “allogeneic” T cell mediated damage but also protect from subsequent syngeneic T cell-mediated pathogenesis that could contribute to prolonged disease. This effect is tissue specific and could in part be due to parenchymal expression of PD-L1 in certain organs. It is possible that such mechanisms could explain more chronic phases of GVHD, which differs from acute GVHD. Ongoing depletion experiments will determine the relative contributions of donor TS1 T cells, donor bone marrow derived T cells and host T cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1094-1094
Author(s):  
Ji Young Lim ◽  
Sung-Eun Lee ◽  
Yoo-Jin Kim ◽  
Gyeongsin Park ◽  
Eun Young Choi ◽  
...  

Abstract Allogeneic hematopoietic stem cell transplantation is an important therapeutic modality used to treat malignancies of hematopoietic origin, such as leukemia and lymphoma. However, development of graft-versus-host disease (GVHD) causes non-relapse mortality and substantial morbidity of recipients. Myeloid differentiation factor 88 (MyD88), a major adaptor mediating TLR signaling, is also known to deliver pro-inflammatory signals. Activation of inflammatory signaling through MyD88 plays a key role in the expansion of myeloid-derived suppressor cells (MDSC) which are a heterogeneous population of immature myeloid ells with anti-inflammatory activity. To explore the contribution of MyD88 expressed by donor bone marrow (BM) cells to development of GVHD, we induced GVHD using T-cell-depleted BM (TCD-BM) isolated from MyD88-deficient (MyD88KO) mice and T cells isolated from wild-type (WT) mice. We employed C57BL/6 (H-2b) → B6D2F1 (H-2b/d) mouse model of GVHD, which differ at major and minor histocompatibility loci. Lethally irradiated B6D2F1 recipient mice were transplanted with either T cell-depleted bone marrow (TCD BM, 5 x 106) from either WT or MyD88KO mice together with WT spleen T cells (1 x 106). Transplantation with MyD88KO TCD BM aggravated GVHD; serious gut damage was evident, with infiltration of T cells specifically into the intestines of recipients. GVHD hosts transplanted with MyD88KO TCD BM exhibited markedly reduced expansion of MDSC. GVHD aggravation after transplantation with MyD88KO TCD-BM, associated with high-level T cell infiltration into the intestine and insignificant expansion of MDSC, was reproduced in another minor histocompatibility mismatch model (C57BL/6 → BALB.B). We next examined allogeneic T cells in the spleens of GVHD hosts in terms of the expression levels of CCR9, which are known to be associated with T cell migration to the intestinal mucosa and the proportion of CCR9 positive cells in CFSE low CD8+ T cells was higher in recipients of MyD88KO TCD BM than WT controls. In parallel, the levels of CCL25 were more highly expressed in the gut of MyD88KO recipients than WT controls. Mixed leukocyte cultures of CFSE- labeled C57BL/6 T cells and irradiated B6DF1 feeder cells were prepared in the presence of MDSC isolated from MyD88KO or WT mice. T cells, co-incubated with MDSC isolated from MyD88KO BM, exhibited a greater extent of CFSE dilution and less Annexin V staining, compared to T cells co-incubated with cells from WT BM. Moreover, MDSC from recipients of MyD88KO TCD BM exhibited a reduced suppressive function, compared to their WT counterparts. Next, we determined whether insufficient expansion of and ineffective suppression by MDSC caused severe GVHD in recipients of MyD88KO TCD BM. Supplementation of transplanted mice with MDSC from WT mice, not from MyD88KO mice, attenuated the severity of GVHD and reduced intestinal T cell infiltration in recipients of MyD88KO TCD BM. To verify the importance of MyD88-mediated signaling by MDSC in protection against severe GVHD, we determined if transplantation with TCD-BM cells containing high levels of MDSC attenuated the severity of GVHD. Pre-treatment of BM donors with lipopolysaccharide increased the frequencies of MDSC and the amounts of MyD88 transcripts in TCD-BM transplant, and alleviated the severity of GVHD and intestinal T-cell infiltration. To explore whether MDSC expansion levels could be used to predict the severity of intestinal GVHD, the T/MDSC ratios were calculated in blood of patients at the time of engraftment and were significantly higher in patients with intestinal GVHD ≥ grade 2. In conclusion, we have shown that MyD88-dependent MDSC expansion from donor BM is critical for protection against fatal acute intestinal GVHD. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document