scholarly journals Veenstra RG, Flynn R, Kreymborg K, et al. B7-H3 expression in donor T cells and host cells negatively regulates acute graft-versus-host disease lethality. Blood. 2015;125(21):3335-3346.

Blood ◽  
2016 ◽  
Vol 127 (24) ◽  
pp. 3104-3104
Blood ◽  
2015 ◽  
Vol 125 (21) ◽  
pp. 3335-3346 ◽  
Author(s):  
R. G. Veenstra ◽  
R. Flynn ◽  
K. Kreymborg ◽  
C. McDonald-Hyman ◽  
A. Saha ◽  
...  

2001 ◽  
Vol 194 (10) ◽  
pp. 1433-1440 ◽  
Author(s):  
Pavan Reddy ◽  
Takanori Teshima ◽  
Mark Kukuruga ◽  
Rainer Ordemann ◽  
Chen Liu ◽  
...  

Interleukin (IL)-18 is a recently discovered cytokine that modulates both T helper type 1 (Th1) and Th2 responses. IL-18 is elevated during acute graft-versus-host disease (GVHD). We investigated the role of IL-18 in this disorder using a well characterized murine bone marrow transplantation (BMT) model (B6 → B6D2F1). Surprisingly, blockade of IL-18 accelerated acute GVHD-related mortality. In contrast, administration of IL-18 reduced serum tumor necrosis factor (TNF)-α and lipopolysaccharide (LPS) levels, decreased intestinal histopathology, and resulted in significantly improved survival (75 vs. 15%, P < 0.001). Administration of IL-18 attenuated early donor T cell expansion and was associated with increased Fas expression and greater apoptosis of donor T cells. The administration of IL-18 no longer protected BMT recipients from GVHD when Fas deficient (lpr) mice were used as donors. IL-18 also lost its ability to protect against acute GVHD when interferon (IFN)-γ knockout mice were used as donors. Together, these results demonstrate that IL-18 regulates acute GVHD by inducing enhanced Fas-mediated apoptosis of donor T cells early after BMT, and donor IFN-γ is critical for this protective effect.


2020 ◽  
Author(s):  
Weijiang Liu ◽  
Na Zhou ◽  
Peng Wang ◽  
Yuanlin Liu ◽  
Wei Zhang ◽  
...  

Abstract Background Mesenchymal stem cells (MSCs) have been utilized in treating acute graft-versus-host disease (aGvHD) as they show strong immunosuppressive capacity, but the mechanisms are not well defined.Methods In this study, we demonstrated that microRNA-223 (miR-223) derived from exosomes secreted by human umbilical cord mesenchymal stem cells (huc-MSCs) and murine compact bone mesenchymal stem cells (mb-MSCs) could inhibit aGvHD progression by reducing the migration and homing of donor T cells in aGvHD mice.Results MiR-223 was one of the conserved microRNAs highly expressed in huc-MSCs exosomes and mMSCs exosomes, which was identified by high-throughput sequencing. MiR-223 derived from MSC exosomes showed enhanced immunosuppressive capacity, as it could inhibit expression of the target gene ICAM-1 and restrain adhesion and migration of T cells in vitro. Moreover, miR-223Agomir was effective in reducing the inflammatory reaction, and declining the donor T cells infiltration into the spleen, liver and intestine in aGvHD mice. Subsequently, it could alleviate aGvHD symptoms. Taken together, the MSC exosome derived miR-223 could attenuate aGvHD in mice through regulating ICAM-1 expression.Conclusions Our results unveil a new role for MSC exosomes derived miR-223 in the treatment of aGvHD.


2007 ◽  
Vol 35 (2) ◽  
pp. 274-286 ◽  
Author(s):  
Yoshinobu Maeda ◽  
Isao Tawara ◽  
Takanori Teshima ◽  
Chen Liu ◽  
Daigo Hashimoto ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (26) ◽  
pp. 2763-2774 ◽  
Author(s):  
Jennifer J. Tsai ◽  
Enrico Velardi ◽  
Yusuke Shono ◽  
Kimon V. Argyropoulos ◽  
Amanda M. Holland ◽  
...  

Abstract Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a ubiquitously expressed transcription factor that is well known for its role in regulating the cellular redox pathway. Although there is mounting evidence suggesting a critical role for Nrf2 in hematopoietic stem cells and innate leukocytes, little is known about its involvement in T-cell biology. In this study, we identified a novel role for Nrf2 in regulating alloreactive T-cell function during allogeneic hematopoietic cell transplantation (allo-HCT). We observed increased expression and nuclear translocation of Nrf2 upon T-cell activation in vitro, especially in CD4+ donor T cells after allo-HCT. Allo-HCT recipients of Nrf2−/− donor T cells had significantly less acute graft-versus-host disease (GVHD)-induced mortality, morbidity, and pathology. This reduction in GVHD was associated with the persistence of Helios+ donor regulatory T cells in the allograft, as well as defective upregulation of the gut-homing receptor LPAM-1 on alloreactive CD8+ T cells. Additionally, Nrf2−/− donor CD8+ T cells demonstrated intact cytotoxicity against allogeneic target cells. Tumor-bearing allo-HCT recipients of Nrf2−/− donor T cells had overall improved survival as a result of preserved graft-versus-tumor activity and reduced GVHD activity. Our findings characterized a previously unrecognized role for Nrf2 in T-cell function, as well as revealed a novel therapeutic target to improve the outcomes of allo-HCT.


2004 ◽  
Vol 174 (1) ◽  
pp. 551-556 ◽  
Author(s):  
Daigo Hashimoto ◽  
Shoji Asakura ◽  
Sachiko Miyake ◽  
Takashi Yamamura ◽  
Luc Van Kaer ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4534-4534
Author(s):  
Govindarajan Thangavelu ◽  
Asim Saha ◽  
Kazutoshi Aoyama ◽  
Cameron McDonald-Hyman ◽  
Yu-Chi Lee ◽  
...  

Abstract Retinoic acid (RA), a metabolite of vitamin A, modulates a variety of aspects of the immune system, primarily because of its diverse effects on a wide range of immune cells. Initiation of RA-mediated transcription requires the binding of RA to heterodimeric nuclear receptors composed of RA receptors (RARα, β, and γ) and retinoid X receptors (RXRα, β, and γ). Although RA signaling is tolerogenic under steady state conditions, previous studies, including ours, have shown that RA can enhance pro-inflammatory responses in acute graft-versus-host disease (aGVHD) (Blood. 2013; 122(12): 2125). In that study, we demonstrated that donor T cells expressing a dominant negative RARα (DNRARα) markedly impaired GVHD lethality capacity. This paradoxical function in aGVHD was attributed to excessive RA production caused by the upregulation of RA-synthesizing enzymes, retinaldehyde dehydrogenases (RALDH), in hematopoietic and non-hematopoietic cells. In the current study, we investigated genetic and translational approaches to ablate RA synthesis and signaling as a means to treat aGVHD. The 4 isoforms of the RALDH enzyme are differentially expressed in a variety of cell types. RALDH-2 is the predominant isoform in dendritic cells (DCs), whereas RALDH-1 levels were found to be higher in intestinal epithelial cells (IECs) during aGVHD. We hypothesized that conditional deletion of either RALDH-2 in host CD11c+ DCs, or RALDH-1 in host IECs using a Cre-Lox system would diminish RA synthesis and reduce aGVHD. Consistent with this hypothesis, ablation of RA synthesis by RALDH-2 in host CD11c+ DCs significantly reduced aGVHD (Figure 1A; p <0.0001) in a fully MHC mismatched BALB/c (H-2d) into C57BL/6 (B6, H-2b) mouse model of aGVHD. However, deletion of RALDH-1 in IECs failed to attenuate aGVHD (not shown), which could be attributed to the existence of other isoforms in these cells. To circumvent the potential contribution of multiple RA synthesizing isoforms, we generated mice over expressing CYP26A1, a key RA catabolizing enzyme. Over expression of CYP26A1 in host hematopoietic cells using CYP26A1stop/stop-VAV-Cre recipients significantly mitigated aGVHD (p<0.005; not shown), providing novel insights into the tissue-specific role of RA signaling in aGVHD. We further explored RA signaling in aGVHD using a translational approach. IRX4204 is a novel and highly specific RXR agonist currently in clinical trials for autoimmune diseases, which can sequester RXR receptors and phenocopies the genetic approach using DNRARα T cells and allowing translation into the clinic. We discovered that IRX4204 can enhance in vitro CD4+CD25+Foxp3+ Regulatory T cell (iTreg) generation from naïve CD4+Foxp3- precursors (Figure 1B), suggesting it might increase Treg in vivo, and therefore be therapeutically useful for aGVHD prevention. To determine the effects of IRX4204 in aGVHD, we used two fully MHC mismatched mouse models: (B6 (H-2b) into BALB/c (H-2d) and BALB/c (H-2d) into B6 (H-2b). Recipients treated with IRX4204 had significantly prolonged survival time (Figure 1C&D), reduced weight loss, and better clinical scores compared to vehicle-treated mice. IRX4204 also significantly reduced donor T cell proliferation, effector differentiation and a 2-4 fold reduced production of pro-inflammatory cytokine (IFN-γ, TNF-α). Despite up-regulating gut-homing receptors on donor T cells, intestinal (and liver) GVHD pathology was reduced, which was associated with higher IEC integrity, assessed by po FITC-dextran serum levels. Compared to controls, IRX4204-treated recipients had a higher frequency of Treg in the spleen, mesenteric lymph nodes (p <0.0001) and colon (p <0.001). To determine whether IRX4204 mitigated aGVHD by promoting peripheral Treg (pTreg) generation, we utilized Treg-depleted donor T cells from either wild type (WT) or Scurfy (Sc) mice in our B6 into BALB/c GVHD model. Sc mice have a FoxP3 gene deletion and therefore cannot produce pTreg. Interestingly, IRX4204 attenuated aGVHD only in recipients that were given WT T cells, indicating that the generation of pTregs was critical for IRX4204-mediated aGVHD protection. Overall, these data lay a strong foundation for the development of novel drug based therapies for aGVHD and other inflammatory disorders by ablating RA synthesis and signaling. Disclosures Chandraratna: Io Pharmaceuticals: Other: Employed by Io Pharmaceuticals and is a board member, holding the titles of title of President and Chief Scientific Officer, and has ownership interest in Io Pharmaceuticals. .


Blood ◽  
2020 ◽  
Vol 136 (4) ◽  
pp. 418-428 ◽  
Author(s):  
Geoffrey R. Hill ◽  
Motoko Koyama

Abstract Allogeneic hematopoietic stem cell transplantation (alloSCT) is an important curative therapy for high-risk hematological malignancies, but the development of severe and/or steroid-refractory acute graft-versus-host disease (aGVHD) remains a significant limitation to optimal outcomes. New approaches to prevent and treat aGVHD remain an unmet need that can be best addressed by understanding the complex disease pathophysiology. It is now clear that chemoradiotherapy used prior to alloSCT induces the release of endogenous alarmins (eg, HMGB-1, ATP, IL-1α, IL-33) from recipient tissue. Exogenous pathogen-derived molecules (eg, lipopolysaccharide, nucleic acids) also translocate from the gastrointestinal tract lumen. Together, these danger signals activate antigen-presenting cells (APCs) to efficiently present alloantigen to donor T cells while releasing cytokines (eg, interleukin-12 [IL-12], IL-23, IL-6, IL-27, IL-10, transforming growth factor-β) that expand and differentiate both pathogenic and regulatory donor T cells. Concurrent costimulatory signals at the APC–T-cell interface (eg, CD80/CD86-CD28, CD40-CD40L, OX40L-OX40, CD155/CD112-DNAM-1) and subsequent coinhibitory signals (eg, CD80/CD86-CTLA4, PDL1/2-PD1, CD155/CD112-TIGIT) are critical to the acquisition of effector T-cell function and ensuing secretion of pathogenic cytokines (eg, IL-17, interferon-γ, tissue necrosis factor, granulocyte-macrophage colony-stimulating factor) and cytolytic degranulation pathway effectors (eg, perforin/granzyme). This review focuses on the combination of cytokine and costimulatory networks at the T-cell surface that culminates in effector function and subsequent aGVHD in target tissue. Together, these pathways now represent robust and clinically tractable targets for preventing the initiation of deleterious immunity after alloSCT.


2021 ◽  
Vol 12 ◽  
Author(s):  
Huihui Liu ◽  
Zhengyu Yu ◽  
Bo Tang ◽  
Shengchao Miao ◽  
Chenchen Qin ◽  
...  

Acute graft-versus-host disease (aGVHD) is a lethal complication after allogeneic hematopoietic stem cell transplantation. The mechanism involves the recognition of host antigens by donor-derived T cells which induces augmented response of alloreactive T cells. In this study, we characterized the role of a previously identified novel classical secretory protein with antitumor function-LYG1 (Lysozyme G-like 1), in aGVHD. LYG1 deficiency reduced the activation of CD4+ T cells and Th1 ratio, but increased Treg ratio in vitro by MLR assay. By using major MHC mismatched aGVHD model, LYG1 deficiency in donor T cells or CD4+ T cells attenuated aGVHD severity, inhibited CD4+ T cells activation and IFN-γ expression, promoted FoxP3 expression, suppressed CXCL9 and CXCL10 expression, restrained allogeneic CD4+ T cells infiltrating in target organs. The function of LYG1 in aGVHD was also confirmed using haploidentical transplant model. Furthermore, administration of recombinant human LYG1 protein intraperitoneally aggravated aGVHD by promoting IFN-γ production and inhibiting FoxP3 expression. The effect of rhLYG1 could partially be abrogated with the absence of IFN-γ. Furthermore, LYG1 deficiency in donor T cells preserved graft-versus-tumor response. In summary, our results indicate LYG1 regulates aGVHD by the alloreactivity of CD4+ T cells and the balance of Th1 and Treg differentiation of allogeneic CD4+ T cells, targeting LYG1 maybe a novel therapeutic strategy for preventing aGVHD.


Sign in / Sign up

Export Citation Format

Share Document