Superagonist CD28 Antibody Preferentially Expanded Foxp3-Expressing nTreg Cells and Prevented Graft-Versus-Host Diseases

2009 ◽  
Vol 18 (5-6) ◽  
pp. 627-638 ◽  
Author(s):  
Yusuke Kitazawa ◽  
Masayuki Fujino ◽  
Xiao-Kang Li ◽  
Lin Xie ◽  
Naotsugu Ichimaru ◽  
...  

Regulatory lymphocytes play a pivotal role in preventing organ-specific autoimmune disease and in induction and maintenance of tolerance in various experimental transplantation models. The enhancement of the number and activity of peripheral CD4+CD25+ Treg cells is an obvious goal for the treatment of autoimmunity and for the suppression of alloreactions. The present study demonstrates that naturally occurring CD4+CD25+ Treg (nTreg) cells preferentially proliferate to a fourfold increase within 3 days in response to the administration of a single superagonistic CD28-specific monoclonal antibody (supCD28 mAb). The appearance of increased Foxp3 molecules was accompanied with polarization toward a Th2 cytokine profile with decreased production of IFN-γ and increased production of IL-4 and IL-10 in the expanded Treg subset. Adoptive transfer of supCD28 mAb-expanded cells in a graft-versus-host disease (GvHD) model induced a potent inhibition of lethality. These results suggest that this therapeutic effect is mediated by the in vivo expansion of nTreg cells. Taken together, these data demonstrate that supCD28-mAb may target nTreg cells in vivo and maintain and enhance their potent regulatory functions for the treatment GvHD.

Blood ◽  
2010 ◽  
Vol 115 (9) ◽  
pp. 1669-1677 ◽  
Author(s):  
Sheng F. Cai ◽  
Xuefang Cao ◽  
Anjum Hassan ◽  
Todd A. Fehniger ◽  
Timothy J. Ley

Abstract Regulatory T (Treg) cells can suppress a wide variety of immune responses, including antitumor and alloimmune responses. The mechanisms by which Treg cells mediate their suppressive effects depend on the context of their activation. We previously reported that granzyme B is important for Treg cell–mediated suppression of antitumor immune responses. We therefore hypothesized that granzyme B may likewise be important for suppression of graft-versus-host disease (GVHD). We found that allogeneic mismatch induces the expression of granzyme B in mixed lymphocyte reactions and in a model of graft-versus-host disease (GVHD). However, wild-type and granzyme B–deficient Treg cells were equally able to suppress effector T (Teff) cell proliferation driven by multiple stimuli, including allogeneicantigen-presenting cells. Surprisingly, adoptive transfer of granzyme B–deficient Treg cells prevented GVHD lethality, suppressed serum cytokine production in vivo, and prevented target organ damage. These data contrast strikingly with our previous study, which demonstrated that granzyme B plays a nonredundant role in Treg cell–mediated suppression of antitumor responses. Taken together, these findings suggest that targeting specific Treg cell–suppressive mechanisms, such as granzyme B, may be therapeutically beneficial for segregating GVHD and graft-versus-tumor immune responses.


Blood ◽  
2008 ◽  
Vol 112 (5) ◽  
pp. 2129-2138 ◽  
Author(s):  
Dongchang Zhao ◽  
Chunyan Zhang ◽  
Tangsheng Yi ◽  
Chia-Lei Lin ◽  
Ivan Todorov ◽  
...  

Abstract CD103 (αEβ7) has been shown to be an excellent marker for identifying in vivo–activated FoxP3+CD4+ regulatory T (Treg) cells. It is unknown whether reinfusion of in vivo–activated donor-type CD103+ Treg cells from recipient can ameliorate ongoing chronic graft-versus-host disease (GVHD). Here, we showed that, in a chronic GVHD model of DBA/2 (H-2d) donor to BALB/c (H-2d) recipient, donor-type CD103+ Treg cells from recipients were much more potent than CD25hi natural Treg cells from donors in reversing clinical signs of GVHD and tissue damage. Furthermore, in contrast to CD25hi natural Treg cells, CD103+ Treg cells expressed high levels of CCR5 but low levels of CD62L and directly migrated to GVHD target tissues. In addition, the CD103+ Treg cells strongly suppressed donor CD4+ T-cell proliferation; they also induced apoptosis of in vivo–activated CD4+ T and B cells and significantly reduced pathogenic T and B cells in GVHD target tissues. These results indicate that CD103+ Treg cells from chronic GVHD recipients are functional, and reinfusion of the CD103+ Treg cells can shift the balance between Treg cells and pathogenic T cells in chronic GVHD recipients and ameliorate ongoing disease.


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 ◽  
2005 ◽  
Vol 105 (5) ◽  
pp. 2220-2226 ◽  
Author(s):  
Joerg Ermann ◽  
Petra Hoffmann ◽  
Matthias Edinger ◽  
Suparna Dutt ◽  
Francis G. Blankenberg ◽  
...  

AbstractCD4+CD25+ regulatory T (Treg) cells are potent modulators of alloimmune responses. In murine models of allogeneic bone marrow transplantation, adoptive transfer of donor CD4+CD25+ Treg cells protects recipient mice from lethal acute graft-versus-host disease (aGVHD) induced by donor CD4+CD25- T cells. Here we examined the differential effect of CD62L+ and CD62L- subsets of CD4+CD25+ Treg cells on aGVHD-related mortality. Both subpopulations showed the characteristic features of CD4+CD25+ Treg cells in vitro and did not induce aGVHD in vivo. However, in cotransfer with donor CD4+CD25- T cells, only the CD62L+ subset of CD4+CD25+ Treg cells prevented severe tissue damage to the colon and protected recipients from lethal aGVHD. Early after transplantation, a higher number of donor-type Treg cells accumulated in host mesenteric lymph node (LN) and spleen when CD4+CD25+CD62L+ Treg cells were transferred compared with the CD62L- subset. Subsequently, CD4+CD25+CD62L+ Treg cells showed a significantly higher capacity than their CD62L- counterpart to inhibit the expansion of donor CD4+CD25- T cells. The ability of Treg cells to efficiently enter the priming sites of pathogenic allo-reactive T cells appears to be a prerequisite for their protective function in aGVHD.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4037-4037
Author(s):  
Lucrezia Colonna ◽  
Mareike Florek ◽  
Dennis B Leveson-Gower ◽  
Emanuela I Sega ◽  
Robert Zeiser ◽  
...  

Abstract Abstract 4037 Allogeneic hematopoietic cell transplantation (HCT) is an effective treatment for many hematologic malignancies such as leukemia, lymphoma and myelodysplastic syndromes. A potentially fatal complication of HCT is graft-versus-host disease (GVHD), where conventional T cells of alloreactive-specificities (CD4+ and CD8+ T cells; Tcon) infiltrate and destroy target organs such as the liver, gut and skin. While immunomodulation is necessary to control the adverse GVHD reactions, an effective immune response is required for successful tumor eradication, named graft-versus-tumor effect (GVT). Following allogeneic bone marrow transplantation, our and other laboratories have shown that the transfer of highly purified regulatory T cells (Treg), a key immunoregulatory cell population involved in the maintenance of immune tolerance, can prevent lethal GVHD induced by the donor-derived Tcon in multiple models across both major and minor histocompatibility barriers. Notably, when transferred into recipient mice with established leukemia or lymphoma, Treg cells effectively suppress Tcon cell proliferation and prevent lethal GVHD, while preserving GVT activity. We have recently adapted our pre-clinical model to an ongoing clinical trial, and are currently assessing the efficacy of highly purified human Treg cells in a clinical setting. However, recent evidence suggest that, under the influence of intense inflammatory signals, the Tregsuppressive phenotype can become unstable, a phenomenon that can culminate in Tregconversion into IL-17-producing pro-inflammatory cells. IL-17 is a proinflammatory cytokine that has been reported to promote GVHD in different models for human HCT. We hypothesized that under the influence of the intense proinflammatory signals, such as those released during an ongoing alloreaction, a fraction of the transferred Treg might be redirected to the Th17 cell fate, thereby losing immunosuppressive potential and acquiring proinflammatory capacity. Our preliminary in vitro studies demonstrated that Treg cells do indeed upregulate Il17 gene expression following activation by allogeneic antigen presenting cells (APCs). We, therefore, sought to evaluate the impact of Il17 gene ablation on Treg stability and immunosuppressive capacity in a major MHC mismatch model, in order to ultimately assess the possible therapeutic potential of IL-17 targeting for the enhancement of Treg-mediated GVHD suppressive ability following HCT. On the basis of our preliminary studies we anticipated that Treg cells purified from Il17 knockout (KO) donor mice would have enhanced immunomodulatory capacity compared to wild-type (WT) Treg, given their inability to produce IL-17. To monitor ongoing GVHD our laboratory has developed an imaging system that allows for the in vivo visualization of T cell homing and proliferation over time, without the necessity to sacrifice the experimental animals. Specifically, the allogeneic Tcon cells are isolated from donor mice where the gene coding for the enzyme luciferase (luc) is expressed by all hematopoietic cells. Importantly, the proliferation of the transferred luc+ Tcon can be directly assessed by the number of photons detected by the bioluminescent imaging system (BLI), which gives a measure of ongoing GVHD. Treg were purified from both WT and IL-17 KO animals and their GVHD-suppressive ability was directly compared in vivo following the transfer of luc+ Tcon. IL-17 KO Tregcells were as proficient as WT Treg in suppressing GVHD. Similarly, the GVHD-inhibitory capacity of Treg cells isolated from IL-1R KO mice, that were previously shown to be resistant to Th17 conversion, was comparable to that of WT Treg. Furthermore, upon ex vivo re-isolation of Tregthat were previously transferred into transplanted recipients, we could detect little or no IL-17 production by intracellular cytokine staining. Thus, we conclude that freshly isolated and highly purified Treg do not convert into IL-17 producing cells in our model, suggesting that IL-17 production is not a factor affecting Tregstability during ongoing GVHD, and excluding IL-17 targeting as a valuable strategy to improve Treg immunotherapy following bone marrow transplantation. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 ◽  
Author(s):  
Syamal K. Datta

Autoantigen-directed tolerance can be induced by certain nucleosomal histone peptide epitope/s in nanomolar dosage leading to sustained remission of disease in mice with spontaneous SLE. By contrast, lupus is accelerated by administration of intact (whole) histones, or whole nucleosomes in microparticles from apoptotic cells, or by post-translationally acetylated histone-peptides. Low-dose therapy with the histone-peptide epitopes simultaneously induces TGFβ and inhibits IL-6 production by DC in vivo, especially pDC, which then induce CD4+CD25+ Treg and CD8+ Treg cells that suppress pathogenic autoimmune response. Both types of induced Treg cells are FoxP3+ and act by producing TGFβ at close cell-to-cell range. No anaphylactic adverse reactions, or generalized immunosuppression have been detected in mice injected with the peptides, because the epitopes are derived from evolutionarily conserved histones in the chromatin; and the peptides are expressed in the thymus during ontogeny, and their native sequences have not been altered. The peptide-induced Treg cells can block severe lupus on adoptive transfer reducing inflammatory cell reaction and infiltration in the kidney. In Humans, similar potent Treg cells are generated by the histone peptide epitopes in vitro in lupus patients’ PBMC, inhibiting anti-dsDNA autoantibody and interferon production. Furthermore, the same types of Treg cells are generated in lupus patients who are in very long-term remission (2-8 years) after undergoing autologous hematopoietic stem cell transplantation. These Treg cells are not found in lupus patients treated conventionally into clinical remission (SLEDAI of 0); and consequently they still harbor pathogenic autoimmune cells, causing subclinical damage. Although antigen-specific therapy with pinpoint accuracy is suitable for straight-forward organ-specific autoimmune diseases, Systemic Lupus is much more complex. The histone peptide epitopes have unique tolerogenic properties for inhibiting Innate immune cells (DC), T cells and B cell populations that are both antigen-specifically and cross-reactively involved in the pathogenic autoimmune response in lupus. The histone peptide tolerance is a natural and non-toxic therapy suitable for treating early lupus, and also maintaining lupus patients after toxic drug therapy. The experimental steps, challenges and possible solutions for successful therapy with these peptide epitopes are discussed in this highly focused review on Systemic Lupus.


2013 ◽  
Vol 150 (3) ◽  
pp. 1024-1031 ◽  
Author(s):  
Mohammad Hossein Boskabady ◽  
Sakine Shahmohammadi Mehrjardi ◽  
Abadorrahim Rezaee ◽  
Houshang Rafatpanah ◽  
Sediqeh Jalali

2021 ◽  
Vol 14 (1) ◽  
pp. 38
Author(s):  
Hyo Jeong Lee ◽  
Pyeonghwa Jeong ◽  
Yeongyu Moon ◽  
Jungil Choi ◽  
Jeong Doo Heo ◽  
...  

Rearranged during transfection (RET), a receptor tyrosine kinase, is activated by glial cell line-derived neurotrophic factor family ligands. Chromosomal rearrangement or point mutations in RET are observed in patients with papillary thyroid and medullary thyroid carcinomas. Oncogenic alteration of RET results in constitutive activation of RET activity. Therefore, inhibiting RET activity has become a target in thyroid cancer therapy. Here, the anti-tumor activity of a novel RET inhibitor was characterized in medullary thyroid carcinoma cells. The indirubin derivative LDD-2633 was tested for RET kinase inhibitory activity. In vitro, LDD-2633 showed potent inhibition of RET kinase activity, with an IC50 of 4.42 nM. The growth of TT thyroid carcinoma cells harboring an RET mutation was suppressed by LDD-2633 treatment via the proliferation suppression and the induction of apoptosis. The effects of LDD-2633 on the RET signaling pathway were examined; LDD-2633 inhibited the phosphorylation of the RET protein and the downstream molecules Shc and ERK1/2. Oral administration of 20 or 40 mg/kg of LDD-2633 induced dose-dependent suppression of TT cell xenograft tumor growth. The in vivo and in vitro experimental results supported the potential use of LDD-2633 as an anticancer drug for thyroid cancers.


Sign in / Sign up

Export Citation Format

Share Document