In vivo induction of regulatory T-cells by a novel bone-marrow derived cell population in the context of successful allotransplantation

2006 ◽  
Vol 130 (2) ◽  
pp. 209-210
Author(s):  
V.R. Shinde Patil ◽  
K.N. Taylor ◽  
Y.L. Colson
Blood ◽  
2012 ◽  
Vol 119 (24) ◽  
pp. 5898-5908 ◽  
Author(s):  
Renee J. Robb ◽  
Katie E. Lineburg ◽  
Rachel D. Kuns ◽  
Yana A. Wilson ◽  
Neil C. Raffelt ◽  
...  

Abstract FoxP3+ confers suppressive properties and is confined to regulatory T cells (Treg) that potently inhibit autoreactive immune responses. In the transplant setting, natural CD4+ Treg are critical in controlling alloreactivity and the establishment of tolerance. We now identify an important CD8+ population of FoxP3+ Treg that convert from CD8+ conventional donor T cells after allogeneic but not syngeneic bone marrow transplantation. These CD8+ Treg undergo conversion in the mesenteric lymph nodes under the influence of recipient dendritic cells and TGF-β. Importantly, this population is as important for protection from GVHD as the well-studied natural CD4+FoxP3+ population and is more potent in exerting class I–restricted and antigen-specific suppression in vitro and in vivo. Critically, CD8+FoxP3+ Treg are exquisitely sensitive to inhibition by cyclosporine but can be massively and specifically expanded in vivo to prevent GVHD by coadministering rapamycin and IL-2 antibody complexes. CD8+FoxP3+ Treg thus represent a new regulatory population with considerable potential to preferentially subvert MHC class I–restricted T-cell responses after bone marrow transplantation.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2195-2195
Author(s):  
William J. Murphy ◽  
Isabel Bareo ◽  
Alan M. Hanash ◽  
Lisbeth A. Welniak ◽  
Kai Sun ◽  
...  

Abstract While a link between the innate to adaptive immune system has been established, studies demonstrating direct effects of T cells in regulating Natural Killer (NK) cell function have been lacking. Naturally occurring CD4+CD25+ regulatory T cells (Tregs) have been shown to potently inhibit adaptive responses by T cells. We therefore investigated whether Tregs could affect NK cell function in vivo. Using a bone marrow transplantation (BMT) model of hybrid resistance, in which parental (H2d) marrow grafts are rejected by the NK cells of the F1 recipients (H2bxd), we demonstrate that the in vivo removal of host Tregs significantly enhances NK-cell mediated BM rejection. This heightened rejection was mediated by the specific NK cell Ly-49+ subset previously demonstrated to reject the BMC in this donor/host pairing. The depletion of Tregs could also further increase rejection already enhanced by treating recipients with the NK cell activator, poly I:C. Although splenic NK cell numbers were not significantly altered, increased splenic NK in vitro cytotoxic activity was observed from the recovered cells. The regulatory role of Tregs was confirmed in adoptive transfer studies in which transferred CD4+CD25+ Tregs resulted in abrogation of NK cell-mediated hybrid resistance. Thus, Tregs can potently inhibit NK cell function in vivo and their depletion may have therapeutic ramifications with NK cell function in BMT and cancer therapy.


2016 ◽  
Vol 301 ◽  
pp. 18-29 ◽  
Author(s):  
Xiaomei Wang ◽  
Cox Terhorst ◽  
Roland W. Herzog

2017 ◽  
Vol 261 ◽  
pp. 223-233 ◽  
Author(s):  
Stephen C. Balmert ◽  
Cara Donahue ◽  
John R. Vu ◽  
Geza Erdos ◽  
Louis D. Falo ◽  
...  

PLoS ONE ◽  
2015 ◽  
Vol 10 (6) ◽  
pp. e0128373 ◽  
Author(s):  
Martijn J. C. van Herwijnen ◽  
Ruurd van der Zee ◽  
Willem van Eden ◽  
Femke Broere

2019 ◽  
Vol 116 (8) ◽  
pp. 3106-3111 ◽  
Author(s):  
Young-Jun Park ◽  
Heeju Ryu ◽  
Garam Choi ◽  
Byung-Seok Kim ◽  
Eun Sook Hwang ◽  
...  

Expression of ectonucleotidase CD39 contributes to the suppressive activity of Foxp3+ regulatory T cells (Tregs) by hydrolyzing immunogenic ATP into AMP. The molecular mechanism that drives CD39 expression on Tregs remains elusive. We found that tumor-infiltrating Tregs (Ti-Tregs) failed to up-regulate CD39 in mice lacking EBI3 subunit of IL-27 or IL-27Ra. Mixed bone marrow chimera and in vitro studies showed that IL-27 signaling in Tregs directly drives CD39 expression on Ti-Tregs in a STAT1-dependent, but STAT3- and T-bet–independent, manner. Tregs stimulated with IL-27 showed enhanced suppressive activities against CD8+ T cell responses in vitro. Moreover, IL-27Ra–deficient Tregs and STAT1-deficient Tregs were less efficient than WT Tregs in suppressing antitumor immunity in vivo. CD39 inhibition significantly abolished IL-27–induced suppressive activities of Tregs. Thus, IL-27 signaling in Tregs critically contributes to protumorigenic properties of Tregs via up-regulation of CD39.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4112-4112
Author(s):  
Jun Li ◽  
Kenrick Semple ◽  
Jessica Heinrichs ◽  
Anandharaman Veerapathran ◽  
Kelley M.K. Haarberg ◽  
...  

Abstract Abstract 4112 Introduction: Naturally occurring regulatory T cells (nTregs) may prevent graft-versus-host disease (GVHD) while preserving graft-versus-leukemia (GVL) activity. However, clinical application of nTregs has been severely hampered by their scarce availability and non-selective suppression. To overcome these limitations, we took an alternative approach to generate antigen-specific induced Tregs (iTregs), and tested their efficacy and selectivity in the prevention of GVHD in pre-clinical models of bone marrow transplantation (BMT). Methods: We selected HY as target antigen, because it is a naturally processed and ubiquitously expressed minor histocompatibility antigen (miHAg) with a proven role in GVHD and GVL effect. To generate HY-specific iTregs, naïve CD4+CD25− cells were isolated from MHC II-restricted HY-specific transgenic mice, and were stimulated with HY peptide and APCs, in the presence of TGFb and retinoic acid. Using similar protocol, we also generated polyclonal iTregs from CD4+CD25− cells of normal C57BL/6 (B6) mice with allogeneic dendritic cells (DCs). iTregs were isolated by positively selecting CD4+CD25hi cells 6–7 days after generation. Frequency of T cells in recognizing alloantigens was measured using a limited dilution assay. Various MHC-mismatched or matched murine BMT models were used, where polyclonal T cells (Teffs hereafter) were transplanted with donor bone marrow to induce GVHD in myeloablative allogeneic recipients. Results: We first assessed the effect of HY-specific iTregs on GVHD using an MHC II-mismatched B6 ® (B6 × bm12)F1 BMT model, and found that HY-specific iTregs prevented GVHD mortality in male (HY+) but not female (HY−) recipients. On the per-cell basis, HY-specific iTregs were significantly more potent than polyclonal Tregs in the prevention of GVHD. By measuring iTregs and Teffs in spleen and liver of the recipients, we found that HY-specific iTregs expanded extensively and significantly suppressed expansion, activation and infiltration of Teffs in male but not female recipients. To exclude the possibility the observation was model specific, we evaluated the efficacy of HY-specific iTregs and found that those iTregs were highly effective in the prevention of GVHD in two additional BMT models, including one MHC-matched but miHAg-mismatched B6 ® BALB.b model and one haplo-mismatched B6 ® B6D2F1 model. To increase translational potential of our approach, we extended our studies to alloantigen-specific polyclonal iTregs. After 7-day culture in iTreg- generating condition with BALB/c DCs, the frequency of B6 iTregs in recognizing BALB/c alloantigens was increased for 16-fold than that of naïve CD4+CD25− T cells. Furthermore, these iTregs were 64–128 fold more suppressive than nTregs to conventional T-cell response against BALB/c alloantigens. Their highly suppressive activity was antigen-specific, because the same alloreactive iTregs had significant lower suppressive activity to T-cell response against the third-party alloantigens. In vivo using an MHC-mismatched B6 ® BALB/c BMT model, we found that these alloreactive iTregs could effectively prevent GVHD at 2:1 ratio of Teff:Treg, at which polyclonal nTregs had a minimal effect. Conclusion: Using monoclonal HY-specific or polyclonal alloantigen-specific iTregs, these studies demonstrate that antigen-specific iTregs were highly effective in controlling GVHD in an activation-dependent manner. Because iTregs specific for a given miHAg (e.g. HY) can control polyclonal Teffs in response to multiple alloantigens and prevent GVHD in allogeneic recipient, these data also indicate that Tregs may control GVHD through a linked-suppression on Teffs in vivo. In conclusion, the current study presents a promising strategy to generate alloantigen-specific iTregs for effective GVHD prevention in human allogeneic hematopoietic cell transplantation. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 2019 ◽  
pp. 1-15 ◽  
Author(s):  
Yejin Xu ◽  
Xinyue Tang ◽  
Min Yang ◽  
Shengguo Zhang ◽  
Shanshan Li ◽  
...  

Aim. To explore the therapeutic effects and mechanisms of interleukin 10 gene-modified bone marrow-derived dendritic cells (DC-IL10) on liver fibrosis. Methods. In vitro, BMDCs were transfected with lentiviral-interleukin 10-GFP (LV-IL10-GFP) at the MOI of 1 : 40. Then, the phenotype (MHCII, CD80, and CD86) and allo-stimulatory ability of DC-IL10 were identified by flow cytometry, and the levels of IL-10 and IL-12 (p70) secreted into the culture supernatants were quantified by ELISA. In vivo, DC-IL10 was injected into mice with CCl4-induced liver fibrosis through the tail vein. Lymphocytes were isolated to investigate the differentiation of T cells, and serum and liver tissue were collected for biochemical, cytokine, histopathologic, immune-histochemical, and Western blot analyzes. Results. In vitro, the expressions of MHCII, CD80, and CD86 in DC-IL10 were significantly suppressed, allogeneic CD4+T cells incubated with DC-IL10 showed a lower proliferative response, and the levels of IL-10 and IL-12 (p70) secreted into the DC-IL10 culture supernatants were significantly increased and decreased, respectively. In vivo, regulatory T cells (Tregs) were significantly increased, while ALT, AST, and inflammatory cytokines were significantly reduced in the DC-IL10 treatment group, and the degree of hepatic fibrosis was obviously reversed. The TGF-β/smad pathway was inhibited following DC-IL10 treatment compared to the liver fibrosis group. Conclusion. IL-10 genetic modification of BMDCs may maintain DC in the state of tolerance and allow DC to induce T cell hyporesponsiveness or tolerance. DC-IL10 suppressed liver fibrosis by inducing Treg production and inhibiting the TGF-β/smad signaling pathway.


2021 ◽  
pp. 2004973
Author(s):  
Gil‐Ran Kim ◽  
Won‐Ju Kim ◽  
Sangho Lim ◽  
Hong‐Gyun Lee ◽  
Ja‐Hyun Koo ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 302-302
Author(s):  
Vu H. Nguyen ◽  
Courtney Wieland ◽  
Christopher Contag ◽  
Robert S. Negrin

Abstract CD4+CD25+ regulatory T-cells (Treg) have the potential to suppress aberrant immune responses and to regulate peripheral T cell homeostasis. In a murine allogeneic bone marrow transplantation (BMT) model, our laboratory has previously shown that Treg suppress graft-versus-host-disease (GVHD) without abrogating the beneficial graft-versus-tumor immunological effect. However, the mechanisms of immuno-regulation, in particular, the allorecognition properties of Treg, their effects on and interaction with other immune cells, and the sites of suppressive activity, remain unclear. In the current study, we investigate the in vivo trafficking of Treg to better understand how localization may affect their regulatory function. Interpretation of previous studies of Treg localization has been limited by several factors: the need to sacrifice mice does not permit tracking the same set of cells over time; data collected at several arbitrary times on selected tissues do not permit accurate determination of primary and secondary sites of migration, activation, proliferation, and suppression; and CD25 expression on activated CD4+CD25- T cells are difficult to distinguish from Treg. We have developed and characterized a transgenic mouse which constitutively expresses the luciferase gene in all hematologic cells. Treg (1x106 cells) from the spleen and lymph nodes of luc+ transgenic FVB/N (H-2q) mice were co-transplanted into lethally-irradiated (800cGy) Balb/c (H-2d) host along with 5x106 wild-type FVB/N T-cell depleted bone marrow (TCD-BM) cells and 2x106 whole splenocytes, the latter containing approximately 30% T cells (Tconv) which induce GVHD. Upon exposure to the substrate luciferin, the luciferase-expressing Treg emit light which can penetrate through living tissues and is captured by sensitive CCD camera detectors. Bioluminescence imaging (BLI) was performed at various time points established previously by our studies of BLI in GVHD. Within the first 48 hours, Treg localized to the cervical lymph nodes (LN) and the spleen. By day 3, signal is detected in other LN (axillary, mesenteric, inguinal) as well as Peyer’s patches and liver. Signal intensity, measured by photons/second/mouse, significantly increased and peaked on day 4, consistent with the migration and proliferation of Treg to and at these secondary lymphoid organs, respectively. Skin infiltration of Treg is noted on day 6, congruent with a decreased intensity in the spleen, liver, and lymph nodes. A similar pattern of early migration and proliferation of effector immune cells is noted in the GVHD control group, which is transplanted with wild-type FVB/N TCD-BM and luc+ FVB/N whole splenocytes. However, with the GVHD group, the signal intensity continues to increase at all sites. Continued BLI of the Treg group up to day 45 demonstrates persistent strong signal in lymphoid organs and skin sites. Clinically, the Treg group has no significant evidence of GVHD. Chimerism studies on day 45 show complete donor origin, however, lymphoid reconstitution of CD4+ and CD8+ T cells is delayed in the GVHD control group and enhanced in the recipients transplanted with Treg. The above results indicate that in vivo, Treg proliferate and survive long-term. In addition, they co-localize with effector immune cells to secondary lymphoid tissues to positively impact clinical outcomes and lymphoid reconstitution following major-MHC mismatched BMT.


Sign in / Sign up

Export Citation Format

Share Document