scholarly journals Retarded recovery of functional T cell frequencies in T cell-depleted bone marrow transplant recipients

Blood ◽  
1987 ◽  
Vol 70 (4) ◽  
pp. 960-964 ◽  
Author(s):  
JP Daley ◽  
MK Rozans ◽  
BR Smith ◽  
SJ Burakoff ◽  
JM Rappeport ◽  
...  

Abstract We have studied the effect of removing donor T cells by treatment with the monoclonal antibody Leu-1 and complement before marrow transplantation on the regeneration of functionally competent T lymphocytes in the blood at selected times after transplant. Using sensitive limiting-dilution methods that allow us to enumerate helper, cytotoxic, and proliferating T lymphocyte precursors, we report that regeneration of a functional T cell compartment is more severely impaired for the first 180 days after transplantation in those patients given T cell-depleted bone marrow than in recipients of untreated marrow. After this first 6 months, however, patients given T cell- depleted bone marrow had blood T cell frequencies comparable to those observed in patients given untreated marrow. Diminished frequencies of reactive T cells in recipients of depleted marrow could leave them more susceptible to infection or to the recurrence of neoplastic cells.

Blood ◽  
1987 ◽  
Vol 70 (4) ◽  
pp. 960-964
Author(s):  
JP Daley ◽  
MK Rozans ◽  
BR Smith ◽  
SJ Burakoff ◽  
JM Rappeport ◽  
...  

We have studied the effect of removing donor T cells by treatment with the monoclonal antibody Leu-1 and complement before marrow transplantation on the regeneration of functionally competent T lymphocytes in the blood at selected times after transplant. Using sensitive limiting-dilution methods that allow us to enumerate helper, cytotoxic, and proliferating T lymphocyte precursors, we report that regeneration of a functional T cell compartment is more severely impaired for the first 180 days after transplantation in those patients given T cell-depleted bone marrow than in recipients of untreated marrow. After this first 6 months, however, patients given T cell- depleted bone marrow had blood T cell frequencies comparable to those observed in patients given untreated marrow. Diminished frequencies of reactive T cells in recipients of depleted marrow could leave them more susceptible to infection or to the recurrence of neoplastic cells.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1331-1331
Author(s):  
Ying Lu ◽  
Wayne Harris ◽  
Jian-Ming Li ◽  
Edmund K. Waller

Abstract Abstract 1331 Poster Board I-353 Background In contrast to the essential role of host dendritic cells (DC) in the initiation of graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL) reactions, less is known about the effects of donor DC on T cells in these processes. We have previously reported that adding donor BM plasmacytoid DC (pDC) progenitors to a murine graft composed of purified hematopoietic stem cells (HSC) and T-cells increased donor activation and Th1 polarization leading to enhanced GVL activity without increasing GVHD (Li et al. 2007 Blood 110:2181), while larger numbers of human donor pDC were associated with less GVL activity following allogeneic bone marrow transplant (BMT) (Waller et al. 2001 Blood 97:2948). To explore the dissociation of GVHD from GVL we tested the hypothesis that activation of donor T-cells by donor pDC leads to reciprocal induction of indoleamine 2,3-dioxygenase (IDO) expression and immune counter-regulatory activity by donor DC that limits donor T-cell allo-reactivity. Methods pDC precursors were purified by high-speed FACS from un-stimulated BM harvested from wild type (WT) and IDO knock-out (IKO) mice. T-cell proliferation and immune polarization in response to indirect antigen presentation by syngenic DC was measured in mixed lymphocyte reaction (MLR) and by recovery of CFSE-labeled donor T-cells from allogeneic transplant recipients. IDO expression in DC was measured by FACS and intracellular staining using pDC from IKO BM as a negative staining control. FACS-purified 5 × 104 pDC either from WT mice or from IKO mice in combination with 3 × 103 c-kit+ Sca-1+ hematopoietic stem cells (HSC) and 3 × 105 T-cells were transplanted in MHC mismatched C57BL/6→B10.BR model following lethal irradiation. Results FACS-purified lineage−CD11cloCD11b− pDC expressed B220 (72%), CD90 (51%), and CD317 (PDCA-1) (93%), had low levels of MHC-II, partial expression of CD4, and lacked expression of CD24, CD80, CD86 and NK cell or granulocytic markers. IDO expression in purified pDC was up regulated by IFN-γ produced by syngenic T-cells in vitro in one-way MLR. In vivo proliferation of CFSE-labeled donor T-cells was enhanced in mice that received pDC from either WT or IKO mice. Co-transplantation of IKO pDC led to higher proliferation rates of CD8+ T-cells but not CD4+ T-cells compared with the proliferation of corresponding donor T-cell subset co-transplanted with WT DC. The incidence and severity of GVHD (weight loss and GVHD score) were markedly increased in recipients receiving pDC from IKO mice as compared with mice receiving WT pDC. The enhanced GVL activity of donor T-cells induced by transplanted donor WT pDC was abolished when IKO pDC were transplanted into tumor-bearing recipients. Transplanting WT donor pDC led to larger numbers of donor-derived CD4+CD25+Foxp3+ T-reg cells in the spleens of transplant recipients compared with mice receiving IKO pDC (p<0.01) in combination with purified HSC and T-cells. Conclusions Taken together, our data suggest IDO expression in pDC as a critical downstream event that inhibits continued T-cell activation and GVHD. We propose a feedback model in which donor pDC initially induce Th1 polarization of activated donor CD8+ T-cells that secret high levels of IFN-γ. IDO expressed by donor pDC in response to local IFN-γ subsequently induces a counter-regulatory effect including the generation of T-reg and down-modulation of CD8+ T-cell allo-reactivity and proliferation, limiting GVHD while preserving the GVL activity of donor T-cells. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4477-4477
Author(s):  
Kaifeng Lisa Lin ◽  
LeShara M Fulton ◽  
Jonathan Serody

The transcription factor aryl hydrocarbon receptor (AhR) is a cytosolic sensor of numerous small synthetic compounds (xenobiotics) and natural chemicals. Ligand binding of AhR causes a conformational change of the receptor, allowing its translocation to nucleus to regulate an array of genes. Different ligands could induce different sets of genes. AhR was first discovered as the mediator of dioxin toxicity. Its role in immunity was recently extensively investigated and expanded exponentially. Here we investigated the role of AhR in donor T cell-induced graft-versus-host diseases (GvHD) after bone marrow transplant in two MHC-mismatched models. B6D2 or Balb/c recipients were irradiated with one dose (950 rads for B6D2 and 800 rads for Balb/c) one day before transplant. C57BL/6 WT or AhR deficient (AhR KO) T cells were transplanted with T cell-depleted bone marrow to B6D2 or Balb/c, at a dose of three million T cells or five hundred thousand T cells, respectively. At different time points, recipient lymphoid organs or target organs were harvested and used for flow analysis or ELISA. A group of recipient mice were observed for an extended time to establish GvHD scores and survival curves. In B6 to B6D2 model, recipient mice transplanted with AhR KO T cells displayed lower GvHD scores and survived longer than mice transplanted with WT T cells (Figure 1C). We found that AhR KO donor T cells failed to accumulate in lymph nodes and spleen as rapidly as WT T cells as evident on day 1 and day 3 post-transplant (Figure 1A). Further investigation showed that the proliferation and cell death were similar between WT and AhR donor T cells, suggesting decreased migration of AhR KO T cells to lymphoid organs. It has been shown that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced AhR activation leads to the up-regulation of CCR4 and CCR5 in T cells, supporting our data that AhR affects cell migration. We also observed that lower percentage of AhR KO T cells produced IFN-g and IL-17 in lymph nodes and spleen early after transplant, but the differences disappeared at later time points (day 7). Colonic homogenates of AhR KO transplants contained less IFN-g, TNF-a, and IL-1a than WT transplants, which indicated that AhR KO T cells caused less inflammation in the gut (Figure 1B). Gut-associated injury due to inflammation is the major cause of death in this model. However, in B6 to Balb/c model, recipient mice transplanted with AhR KO T cells displayed the opposite survival curve. They had diminished survival compared to mice receiving WT T cells with most mice succumbing to GvHD around day 10 to day 15 before the onset of diarrhea. AhR KO T cells in this model still accumulated less in lymph nodes at early time points, which suggested that AhR played a role in cell migration in both models. It was noteworthy that after day 6, the number of AhR KO T cells in the spleen was actually similar to or higher than WT T cells in both models, suggesting cell proliferation was not affected by AhR deficiency. In conclusion, AhR KO T cells demonstrate decreased migration to lymphoid organs after bone marrow transplant compared to WT T cells. This decrease in migration may be the reason why mice transplanted with AhR KO T cells exhibit less inflammation in the gut and subsequently survive better from GvHD in the B6 to B6D2 model. However, mice transplanted with AhR KO T cells have diminished survival in B6 to Balb/c model despite of lower T cell numbers in lymph nodes at early time points. Thus, certain function of AhR in donor T cells is model dependent while other function is conserved among different models. These data suggest that investigators should be cautious regarding the supplementation of AhR ligands to diminish GvHD in clinical studies. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3511-3511
Author(s):  
Hongmei Li ◽  
Daniel Kaplan ◽  
Anthony Jake Demetris ◽  
Jennifer McNiff ◽  
Mark Shlomchik ◽  
...  

Abstract Graft-versus-host disease (GVHD) is initiated when alloreactive donor T cells are primed by professional antigen-presenting cells (APCs) to undergo clonal expansion and maturation. Host APCs that survive pretransplant conditioning play an essential role in this T cell activation, and are an attractive target for GVHD prevention and treatment. However, APCs are diverse in phenotype, location and function and an understanding of the roles of distinct subsets is an important first step in developing APC-targeted therapies. Skin is the most frequently affected organ in GVHD. Langerhans cells (LCs), characterized by expression of Langerin, are a major APC in the epidermis, and thus it was logical to hypothesize that host LCs would have a role in GVHD induction. Indeed, in an MHC-mismatched model, Merad et al. showed that host LCs persist after T cell-depleted (TCD) but not T cell-replete bone marrow transplant (BMT), and that these host LCs in donor→host chimeras are sufficient to induce skin GVHD after a second allogeneic bone marrow transplant (alloBMT). However, this work did not examine the role of recipient LCs when all other APCs are intact, the scenario at the time of transplant in all patients. To address this question, we created a transgenic mouse that constitutively lacks epidermal LCs. We did so by expressing diphtheria toxin A chain (DTA) driven by the human Langerin gene (Kaplan, et al 2005) in a bacterial artificial chromosome (BAC). We used Langerin-DTA BAC transgene positive (Tg+) mice or Tg-littermates as recipients in the C3H.SW (H-2b)→B6 (H-2b) strain paring, in which recipient APCs are necessary and sufficient for GVHD induction. Tg+ and Tg− CD8 recipients developed similar GVHD as measured by weight loss and clinical skin disease. Tg+ and Tg− CD8 recipients also had comparable pathologic GVHD of the skin, ear, liver and colon. To generalize these findings, we used B6bm12 →B6 strain pairing, an MHCII-mismatched CD4-dependent GVHD model, in which recipient APCs are also required (Teshima et al, 2002). Tg+ and Tg− CD4 recipients developed similar weight loss and pathologic changes in the tongue and liver, primary sites of GVHD in this model. Thus, in both MHC-matched and MHC-mismatched models in which recipient APCs are absolutely required, the specific absence of recipient epidermal LCs did not affect clinical or histological GVHD. We also analyzed LC turnover in these alloBMT recipients. As previously reported, LCs remained host-derived in B6 Tg− recipients of TCD C3H.SW bone marrow. Given our prior result that C3H.SW → B6 chimeras are resistant to GVHD induction by a second alloBMT from C3H. SW donors (Shlomchik, et al 1999), unlike in the MHC-mismatched model employed by Merad, residual host LCs are insufficient to initiate GVHD in this MHC-matched system. In B6 Tg− recipients of TCD C3H.SW bone marrow plus GVHD-inducing CD8 cells, LC turnover varied by mouse and ranged from all host or donor to a mix of donor and host LCs. This variability could relate to the extent of skin GVHD, as we previously found that epidermal MHCII+ cells in skin GVHD lesions in this model are donor-derived (Matte et all, 2004). Strikingly, in contrast to Tg− recipients, donor-derived LCs developed in Tg+ recipients of TCD C3H.SW bone marrow. Donor LCs also engrafted in Tg+ recipients of TCD bone marrow from Tg− but otherwise syngeneic littermates or B6 RAG1−/− T cell-deficient donors. Thus, in contrast to LC-replete mice, neither allogeneic donor T cells nor UV-induced inflammation was required for donor LC engraftment in LC-deficient hosts. These data indicate that a Langerin+ cell, absent in Langerin-DTA Tg+ mice, regulates LC turnover in the absence of inflammation. Work is underway to identify this key cell.


Blood ◽  
2008 ◽  
Vol 112 (6) ◽  
pp. 2232-2241 ◽  
Author(s):  
Jeff K. Davies ◽  
John G. Gribben ◽  
Lisa L. Brennan ◽  
Dongin Yuk ◽  
Lee M. Nadler ◽  
...  

AbstractWe report the outcomes of 24 patients with high-risk hematologic malignancies or bone marrow failure (BMF) who received haploidentical bone marrow transplantation (BMT) after ex vivo induction of alloantigen-specific anergy in donor T cells by allostimulation in the presence of costimulatory blockade. Ninety-five percent of evaluable patients engrafted and achieved full donor chimerism. Despite receiving a median T-cell dose of 29 ×106/kg, only 5 of 21 evaluable patients developed grade C (n = 4) or D (n = 1) acute graft-versus-host disease (GVHD), with only one attributable death. Twelve patients died from treatment-related mortality (TRM). Patients reconstituted T-cell subsets and immunoglobulin levels rapidly with evidence of in vivo expansion of pathogen-specific T cells in the early posttransplantation period. Five patients reactivated cytomegalovirus (CMV), only one of whom required extended antiviral treatment. No deaths were attributable to CMV or other viral infections. Only 1 of 12 evaluable patients developed chronic GVHD. Eight patients survive disease-free with normal performance scores (median follow-up, 7 years). Thus, despite significant early TRM, ex vivo alloanergization can support administration of large numbers of haploidentical donor T cells, resulting in rapid immune reconstitution with very few viral infections. Surviving patients have excellent performance status and a low rate of chronic GVHD.


2019 ◽  
Vol 4 (42) ◽  
pp. eaay8556 ◽  
Author(s):  
S. Takashima ◽  
M. L. Martin ◽  
S. A. Jansen ◽  
Y. Fu ◽  
J. Bos ◽  
...  

Despite the importance of intestinal stem cells (ISCs) for epithelial maintenance, there is limited understanding of how immune-mediated damage affects ISCs and their niche. We found that stem cell compartment injury is a shared feature of both alloreactive and autoreactive intestinal immunopathology, reducing ISCs and impairing their recovery in T cell–mediated injury models. Although imaging revealed few T cells near the stem cell compartment in healthy mice, donor T cells infiltrating the intestinal mucosa after allogeneic bone marrow transplantation (BMT) primarily localized to the crypt region lamina propria. Further modeling with ex vivo epithelial cultures indicated ISC depletion and impaired human as well as murine organoid survival upon coculture with activated T cells, and screening of effector pathways identified interferon-γ (IFNγ) as a principal mediator of ISC compartment damage. IFNγ induced JAK1- and STAT1-dependent toxicity, initiating a proapoptotic gene expression program and stem cell death. BMT with IFNγ–deficient donor T cells, with recipients lacking the IFNγ receptor (IFNγR) specifically in the intestinal epithelium, and with pharmacologic inhibition of JAK signaling all resulted in protection of the stem cell compartment. In addition, epithelial cultures with Paneth cell–deficient organoids, IFNγR-deficient Paneth cells, IFNγR–deficient ISCs, and purified stem cell colonies all indicated direct targeting of the ISCs that was not dependent on injury to the Paneth cell niche. Dysregulated T cell activation and IFNγ production are thus potent mediators of ISC injury, and blockade of JAK/STAT signaling within target tissue stem cells can prevent this T cell–mediated pathology.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1732-1732
Author(s):  
Suigui Wan ◽  
Changqing Xia ◽  
Laurence Morel

Abstract The B6.Sle1.Sle2.Sle3 triple congenic mouse (B6.TC) is a model of lupus due to the co-expression of the three major NZM2410-derived susceptibility loci on a C57BL/6 background. B6.TC mice produce high titers of anti-nuclear nephrogenic autoantibodies and a highly penetrant glomerulonephritis. Previous studies have shown the Sle1 locus is associated with a reduced number of regulatory T cells (Treg), and that Sle3 results in intrinsic defects in myeloid cells that hyperactivate T cells. Here, we show that B6.TC dendritic cells (DCs) accumulate in lymphoid organs and present a defective maturation process, in which bone-marrow derived DCs, plasmacytoid and myeloid DCs express a significantly lower level of CD80, CD86 and class II MHC than B6 controls. B6.TC DCs also induce a higher level of proliferation in CD4+ T cells than B6 DCs, and B6.TC DCs block the suppressive activity of Treg. B6.TC DCs over-produce IL-6, which is necessary for the blockade of Treg activity, as shown by anti-IL-6 neutralizing antibody in the suppression assays. The over-production of IL-6 by DCs and the blockade of Treg activity maps to Sle1, which therefore not only confers a reduced number of Treg, but also blocks their ability to regulate autoreactive T cells. Taken together, these results provide a genetic and mechanistic evidence for systemic autoimmunity resulting from an impaired regulatory T cell compartment both in number and function, and for Sle1-expressing DCs playing a major role in the latter defect though their production of IL-6.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1880-1880
Author(s):  
Trisha Dant ◽  
Danny Bruce ◽  
Leshara Fulton ◽  
Michelle West ◽  
Niko Foger ◽  
...  

Abstract Allogeneic stem cell transplant is a standard treatment for patients with high-risk and relapsed myeloid and lymphoid malignancies. However, donor T cells from the stem cell graft mediate graft-versus-host disease (GVHD), which is a common cause of morbidity and mortality for transplant recipients. Our group and others have shown that migration of donor T cells into secondary lymphoid tissue (SLT) and subsequent migration to target organs is critical to the pathogenesis of acute GVHD. The Coronin family of proteins consists of actin-binding proteins, which regulate filament formation by interacting with the Arp2/3 complex. Coronin 1B, a ubiquitously expressed member of the Coronin family, is required for lamellipodial protrusion and effective cell migration. Previous work has not evaluated a role for this protein in the function of T lymphocytes or during acute GVHD. To evaluate the effect of Coronin 1B in acute GVHD pathogenesis, we transplanted B6 T cell depleted bone marrow cells with wild type or Coronin 1B-/- T cells to lethally irradiated B6D2 and BALB/c recipient mice and evaluated clinical score of GVHD and overall survival. B6D2 recipients of Coronin 1B-/- T cells demonstrated 100% survival (Figure 1A. p< .001 as determined by Log-rank (Mantel-Cox) test) and significantly decreased clinical scores after transplant. This was confirmed with improvement in survival in BALB/c recipients of Coronin 1B-/- T cells. Additionally, Coronin 1B-/- T cells were capable of eliminating P815 tumor cells, indicating that loss of Coronin 1B does not inhibit graft-versus-tumor activity. By day 12 post- transplant, all mice receiving bone marrow alone developed tumor compared to none of the mice receiving Coronin 1B-/- T Cells. However, protection was not complete as 40% of Coronin 1B-/- T cell recipients developed tumor by day 23. To determine the effect of Coronin 1B on T cell migration during GVHD, B6D2 recipients were given GFP-expressing wild type or Coronin 1B-/- T cells along with T cell depleted bone marrow. Lymphoid tissue and target organs were harvested and analyzed by flow cytometry or GFP ELISA. We observed decreased accumulation of Coronin 1B-/- CD4+ (Figure 1B. p< .01 as determined by Student's t -test) and CD8+ T cells in the inguinal lymph node, mesenteric lymph node, and the spleen 4 days after transplant with no difference in accumulation in lymphoid tissue on days 7 and 14 after transplant. Additionally, we found decreased accumulation of Coronin 1B-/- donor T cells in the lung, colon and spleen 14 days after transplant (Figure 1C. p< .05 by Student's t -test). We also quantified the amount of cytokine in target organs by ELISA, and observed a decrease in IFN-γ and TNF-α in the colon 14 days after transplant. Our data demonstrate that Coronin 1B-/- T cells elicit reduced GVHD compared to wild type T cells. This was correlated with decreased accumulation of Coronin 1B-/- T cells in SLT early after transplant. These data indicate that targeting the migration of T cells to SLT is a viable approach to prevent acute GVHD. Figure 1. (A) Kaplan Meier curve comparing B6D2 recipients of Coronin 1B-/- T cells and wild type (WT) T Cells. (B) Decreased accumulation of Coronin 1B-/- T Cells 4 Days after transplant. For panels (B) and (C) black bars indicate recipients of WT T cells while red bars indicate recipients of Coronin 1B-/- T cells. Inguinal lymph nodes (ILN) were pooled from n=5 mice from each group. Spleens were analyzed individually. GFP expressing donor cells were analyzed by flow cytometry. Representative image of two experiments. (C) Coronin 1B-/- T cells express decreased accumulation in the lung, colon and spleen 14 days after transplant. Target organs were analyzed by GFP ELISA to detect GFP+ Donor Cells (n=5 in each group). Figure 1. (A) Kaplan Meier curve comparing B6D2 recipients of Coronin 1B-/- T cells and wild type (WT) T Cells. (B) Decreased accumulation of Coronin 1B-/- T Cells 4 Days after transplant. For panels (B) and (C) black bars indicate recipients of WT T cells while red bars indicate recipients of Coronin 1B-/- T cells. Inguinal lymph nodes (ILN) were pooled from n=5 mice from each group. Spleens were analyzed individually. GFP expressing donor cells were analyzed by flow cytometry. Representative image of two experiments. (C) Coronin 1B-/- T cells express decreased accumulation in the lung, colon and spleen 14 days after transplant. Target organs were analyzed by GFP ELISA to detect GFP+ Donor Cells (n=5 in each group). 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.


Sign in / Sign up

Export Citation Format

Share Document