scholarly journals Inducible T-Cell Co-Stimulator Impacts Chronic Graft-Versus-Host Disease by Regulating Both Pathogenic and Regulatory T Cells

2018 ◽  
Vol 9 ◽  
Author(s):  
Mengmeng Zhang ◽  
Yongxia Wu ◽  
David Bastian ◽  
Supinya Iamsawat ◽  
Jinsam Chang ◽  
...  
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 ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4485-4485
Author(s):  
Antonio Pierini ◽  
Dominik Schneidawind ◽  
Mareike Florek ◽  
Maite Alvarez ◽  
Yuqiong Pan ◽  
...  

Donor derived regulatory T cells (Tregs) effectively prevent graft versus host disease (GVHD) in mouse models and in early phase clinical trials. Interleukin 2 (IL-2) therapy in patients with chronic GVHD (cGVHD) can increase Treg number and the Treg/CD4+ T cell ratio resulting in organ damage reduction and symptom relief. Less is known regarding Treg-based treatment for acute GVHD (aGVHD). In this study we evaluated the role of donor Treg cellular therapy for aGVHD treatment in well established murine models. T cell depleted bone marrow (TCD BM) from C57BL/6 mice was transplanted into lethally irradiated (8 Gy) BALB/C recipients together with 7.5x105 to 1x106/animal donor derived luc+ Tcons. Naturally occurring CD4+CD25+FoxP3+ donor type Tregs (nTregs) were purified from C57BL/6 donor mice. 2.5x105/mouse nTregs were injected at day 6 or 7 after transplant in mice that showed clear clinical signs of aGVHD and Tcon proliferation assessed by bioluminescence imaging (BLI). Survival analysis showed a favorable trend for nTreg treated mice, but the impact of this treatment was modest and not statistically significant (p 0.08). aGVHD is a disease characterized by the activation and rapid proliferation of alloreactive donor conventional T cells (Tcons) directed against host antigens, so one of the major obstacles of this approach is to overcome the large number and effector function of activated Tcons. Several studies have utilized ex vivo expansion of Tregs to increase their number with the goal of maintaining suppressive function. We developed a different strategy with the intent to “educate” Tregs to specifically suppress the reactive Tcon population. We incubated 2.5x105 donor derived Tregs with irradiated (3000 cGy) blood of aGVHD affected mice for 20 hours without further stimulation and injected the entire pool of these cells, termed educated Treg (eTregs), at day 7 or 8 after transplant and Tcon injection. Interestingly eTregs significantly improved aGVHD affected mouse survival (p = 0.0025 vs Tcons alone). BLI showed no difference between the groups (p = 0.85) because the treatment intervened after Tcon proliferation and activation was initiated. To evaluate eTreg impact on graft versus tumor (GVT) effects, we transplanted BALB/C mice with C57BL/6 TCD BM and 1x104/mouse luc+ A20 tumor cells along with 1x106/mouse donor Tcons and 2.5x105 eTregs. Mice that received TCD BM and A20 tumor cells alone died from progressive tumor growth, while mice that received Tcons died from GVHD without tumor engraftment. Further animals that received both Tcon and eTreg treatment did not have tumor engraftment demonstrating that eTregs do not impact Tcon mediated GVT effects. Further studies are ongoing to characterize the eTreg population as compared to nTreg, with respect to expression of activation markers and in functional assays. Our observations indicate that Tregs can be ex vivo educated to suppress in vivo reactive and proliferating Tcons. Moreover our data demonstrate that eTreg adoptive transfer is clinically feasible and promising. These findings may be relevant for the development of clinical grade Treg based cellular therapy for the treatment of conditions caused by immune dysregulation such as aGVHD and autoimmune diseases and for transplant tolerance induction. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 38 (5) ◽  
pp. 494-501 ◽  
Author(s):  
Juyang Kim ◽  
Hye J Kim ◽  
Woon S Choi ◽  
Seok H Nam ◽  
Hong R Cho ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3176-3176
Author(s):  
Robert Zeiser ◽  
Vu H. Nguyen ◽  
Jing-Zhou Hou ◽  
Andreas Beilhack ◽  
Elizabeth A. Zambricki ◽  
...  

Abstract CD4+CD25+ regulatory T cells (Treg) have been demonstrated to reduce the severity of acute graft-versus-host disease (aGvHD) in murine models of bone marrow transplantation. However, the surface molecules that are critical for suppression are unclear. The TNF-R superfamily member CD30 has been shown to be expressed on regulatory T cells that down-modulate nickel specific immune responses and to be relevant for Treg mediated protection from allograft rejection. Deficiency of the CD30 molecule (CD30−/−) is associated with impaired thymic negative selection and augmented T cell autoreactivity. Therefore, we investigated the role of CD30 signaling in Treg function in an aGvHD model. Treg derived from CD30−/ − animals were significantly less effective in preventing aGvHD lethality (wt vs. CD30−/ − p=0.002). Signal intensity derived from expanding luciferase expressing alloreactive conventional T cells (Tconv) was significantly higher if CD30−/ − Treg as compared to wt Treg (p=0.007) were transferred as assessed by bioluminescencent based imaging. Blockade of the CD30/CD153 pathway with a neutralizing anti-CD153 mAb during the early (days −2 to +4) but not late (days +4 to +10) phase of adoptive Treg transfer reduced Treg mediated protection from proinflammatory cytokine accumulation and apoptosis of donor-type CD4 and CD8 T cells. In vivo bioluminescence imaging demonstrated intact Treg homing, but reduced expansion when CD153 was blocked during the early phase after adoptive transfer. CD30 surface expression on Treg increased with alloantigen exposure and CD153 expression on recipient-type dendritic cells increased in the presence of an irradiation induced proinflammatory environment but not when T cell depleted bone marrow and Tconv were transferred into non-irradiated Rag 2−/ −γc−/ − recipients. These data are the first to demonstrate that early CD30 signaling is relevant for Treg mediated aGvHD protection after major MHC mismatch bone marrow transplantation.


Blood ◽  
2009 ◽  
Vol 114 (6) ◽  
pp. 1263-1269 ◽  
Author(s):  
Christian Becker ◽  
Christian Taube ◽  
Tobias Bopp ◽  
Christoph Becker ◽  
Kai Michel ◽  
...  

AbstractNaturally occurring CD4+CD25+ regulatory T cells (Tregs) represent a unique T-cell lineage that is endowed with the ability to actively suppress immune responses. Therefore, approaches to modulate Treg function in vivo could provide ways to enhance or reduce immune responses and lead to novel therapies. Here we show that the CD4 binding human immunodeficiency virus-1 envelope glycoprotein gp120 is a useful and potent tool for functional activation of human Tregs in vitro and in vivo. Gp120 activates human Tregs by binding and signaling through CD4. Upon stimulation with gp120, human Tregs accumulate cyclic adenosine monophosphate (cAMP) in their cytosol. Inhibition of endogeneous cAMP synthesis prevents gp120-mediated Treg activation. Employing a xenogeneic graft versus host disease model that has been shown to be applicable for the functional analysis of human Tregs in vivo, we further show that a single dose of gp120 is sufficient to prevent lethal graft versus host disease and that the tolerizing effect of gp120 is strictly dependent on the presence of human Tregs and their up-regulation of cAMP upon gp120-mediated activation. Our findings demonstrate that stimulation via the CD4 receptor represents a T-cell receptor–independent Treg activating pathway with potential to induce immunologic tolerance in vivo.


Blood ◽  
2006 ◽  
Vol 109 (5) ◽  
pp. 2225-2233 ◽  
Author(s):  
Robert Zeiser ◽  
Vu H. Nguyen ◽  
Jing-Zhou Hou ◽  
Andreas Beilhack ◽  
Elizabeth Zambricki ◽  
...  

Abstract Murine CD4+CD25+ regulatory T cells (Treg cells) reduce acute graft-versus-host disease (aGvHD). However, surface molecules critical for suppression are unclear. Deficiency of CD30 (CD30−/−) leads to impaired thymic negative selection and augmented T-cell autoreactivity. Therefore, we investigated the role of CD30 signaling in Treg-cell function during aGvHD. Treg cells derived from CD30−/− animals were significantly less effective in preventing aGvHD lethality. Early blockade of the CD30/CD153 pathway with a neutralizing anti-CD153 mAb reduced Treg-mediated protection from proinflammatory cytokine accumulation and donor-type T-cell apoptosis. In vivo bioluminescence imaging demonstrated intact homing but reduced expansion of luciferase-expressing Treg cells when CD153 was blocked during the early phase after adoptive transfer. CD30 surface expression on Treg cells increased with alloantigen exposure, and CD153 expression on recipient-type dendritic cells increased in the presence of a proinflammatory environment. These data demonstrate that early CD30 signaling is critical for Treg-mediated aGvHD protection after major MHC-mismatch bone marrow transplantation.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3244-3244
Author(s):  
Hideaki Fujiwara ◽  
Yoshinobu Maeda ◽  
Koichiro Kobayashi ◽  
Hisakazu Nishimori ◽  
Ken-ichi Matsuoka ◽  
...  

Abstract Chronic graft-versus-host disease (cGVHD) remains a major cause of late-phase mortality and morbidity after allogeneic hematopoietic stem cell transplantation. Programmed death-1 (PD-1) and its ligands B7H1 and B7DC, which deliver inhibitory signals and regulate T cell activation, tolerance, and immunopathology, are involved in autoimmune disease. Although several studies have shown that blocking the PD-1 pathway enhances acute GVHD, its relationship to cGVHD remains unclear. We investigated the role of the PD-1 pathway in cGVHD, using a well-defined mouse cGVHD model. Recipients received 6 Gy TBI and were transplanted with purified splenic T cells and BM from either syngeneic BALB/c or allogeneic B10.D2 donors. On days 14, 21, 28, and 56 post-transplant, CD4 and CD8 cells from the spleens and peripheral lymph nodes (pLN) in the allogeneic recipients expressed significantly more PD-1 than those in the syngeneic recipients (p<0.005). Allogeneic recipients had elevated B7H1 mRNA levels from day 14-28 post-transplant (p<0.05) and immunohistochemical analysis of skins from allogeneic recipients showed more B7-H1 expression than from syngeneic recipients on days 14-28 post-transplant, while this declined to the same level as in the syngeneic group after day 42. These findings suggest that donor cells express more PD-1, while target tissues of recipients transiently up-regulate B7H1 expression only in the early phase and soon down-regulate it to syngeneic levels. Upon transfer of PD-1-/- donor T cells with the B10.D2 background into allogeneic BMT models, weight loss was severe and 100% of the recipients died by day 23 post-transplant. To avoid early death, we administered antibodies blocking the PD-1 pathway to recipients of WT donors beginning on day 14 post-transplant, just before they developed clinical signs of cGVHD. Mice treated with anti-PD-1 Ab showed 70% mortality by day 35 and 10% mortality was seen in those given anti-B7H1 Ab or anti-B7DC Ab. All groups treated with anti-PD-1, anti-B7H1, or anti-B7DC Ab had significantly higher cGVHD scores than controls (p<0.05). We next used B7H1-/- mice with the BALB/c background as recipients to evaluate how host B7H1 expression contributes to cGVHD. Allogeneic B7H1-/- recipients showed significantly more severe skin cGVHD and histopathological damage than WT controls (5.86 ± 0.85 vs. 8.38 ± 0.38, p<0.05). We previously elucidated the contribution of Th1+Th17+T cells to cGVHD and Am80, a potent synthetic retinoid, regulated both Th1 and Th17 responses, resulting in an attenuation of cGVHD (Nishimori et al. Blood 2012). Recipients of B7H1-/- showed fewer Foxp3+ regulatory T cells in the early phase (day 14, 13.5±4.2% vs. 6.4±4.4%, p<0.05), whereas there was no difference in the frequency of Foxp3+ regulatory T cells in the late transplantation period (day 28), as compared to WT recipients. Th1+Th17+T cells were detected significantly more frequently in recipients of B7H1-/- donors than those of WT recipients (day 28 2.7±0.35% vs. 1.4±0.2%, p<0.05). Administration of Am80 reduced Th1+Th17+T cells and cGVHD damage in recipients of B7H1-/- donors. To explore the contribution of B7H1 expression on hematopoietic cells or non-hematopoietic cells to cGVHD, (B7H1-/-→WT), (WT→B7H1-/-), and (WT→WT) chimeric mice (BALB/c background) were created by reconstituting sublethally irradiated WT or B7H1-/- Balb/c mice with BM cells from WT or B7H1-/- BALB/c mice. There were no differences in the clinical and pathological cGVHD scores between (B7H1-/-→WT) and (WT→WT) chimeric mice. CD4+CD25+ Foxp3+ Treg cells from (B7H1-/-→WT) recipients were detected less frequently on day 14 than in (WT→WT) recipients (p<0.05), but at similar levels on days 21 and 28. These findings suggest that B7H1 expression on hematopoietic cells plays a role in the development of Tregs only during the early transplantation period, but does not affect cGVHD severity. Unlike (B7H1-/-→WT) recipients, (WT→B7H1-/-) chimeras had significantly worse clinical cGVHD scores (p<0.05), histopathological damage (p<0.05), and Th1+Th17+T cell expansion (p<0.05), but no Treg cell changes. Collectively, these findings indicated that B7H1 expression on host tissues was dedicated to the expansion of IFN-g/IL-17 double-positive cells leading to cGVHD and that modulation of the tissue expression of B7-H1 might represent a new strategy for preventing or treating cGVHD. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document