scholarly journals In vitro expanded human CD4+CD25+ regulatory T cells suppress effector T cell proliferation

2005 ◽  
Vol 115 (1) ◽  
pp. 3-9 ◽  
Author(s):  
K EARLE ◽  
Q TANG ◽  
X ZHOU ◽  
W LIU ◽  
S ZHU ◽  
...  
2007 ◽  
Vol 123 ◽  
pp. S4
Author(s):  
Todd Brusko ◽  
Clive Wasserfall ◽  
Kieran McGrail ◽  
Marcus Moore ◽  
Alyssa Huegel ◽  
...  

2021 ◽  
Author(s):  
◽  
Aras Toker

<p>Glatiramer acetate (GA) is approved for the treatment of relapsing-remitting multiple sclerosis (MS), and can suppress experimental autoimmune encephalomyelitis (EAE), a murine model of human MS. GA treatment is associated with the induction of anti-inflammatory TH2 responses and with the antigen specific expansion of regulatory T cells that counteract or inhibit pathogenic events in MS and EAE. These T cell mediated mechanisms of protection are considered to be a result of modulation of antigen presenting cells (APCs) by GA, rather than direct effects on T cells. However, it is unknown if GA preferentially targets a specific APC subset or can act through multiple APCs in vivo. In addition, GA-modulated innate cells may also exhibit direct antigen non-specific suppression of autoreactive cells. One objective of this study was to identify the in vivo target cell population of GA and to assess the potential of the target cells to antigen non-specifically suppress immune responses. Fluorophor-labelled GA bound to monocytes after intravenous injections, suggesting that monocytes may be the primary target of GA in vivo. In addition, intravenous GA treatment enhanced the intrinsic ability of monocytes to suppress T cell proliferation, both in vitro and in vivo. The findings of this study therefore suggest that GA-induced monocytes may contribute to GA therapy through direct mechanisms of antigen non-specific T cell immunosuppression. A further objective of this work was to investigate the potential of an in vivo drug targeting approach. This approach was hypothesised to increase the uptake of GA by the target cells and substantially improve GA treatment through antigen specific mechanisms such as induction of TH2 or regulatory T cells. Targeting antigens to professional APCs with an anti-MHC class II antibody resulted in significantly enhanced T cell proliferation in vitro. However, no EAE suppression occurred when GA was targeted to MHC class II in vivo. In addition, targeting GA specifically to monocytes also failed to suppress EAE. These findings suggest that GA treatment may selectively modulate monocytes to enhance their ability to inhibit autoreactive T cells, which could be part of the mechanism by which GA ameliorates MS. Targeting GA to a specific cell type may not be a powerful approach to improve treatment, because increased proliferation of GA specific T cells is not sufficient for disease suppression, and conjugation to antibodies may functionally reduce GA to a mere antigen devoid of immunomodulatory capacity.</p>


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1008-1008
Author(s):  
Karnail Singh ◽  
Natalia Kozyr ◽  
Linda Stempora ◽  
Allan D Kirk ◽  
Christian P Larsen ◽  
...  

Abstract Abstract 1008 Regulatory T cells (Tregs) have been shown to be potent inhibitors of autoimmunity, and to be capable of suppressing alloimmune responses that occur during both allograft rejection and graft-versus host disease. However, they have yet to gain widespread use clinically, due in part to the fact that it remains extremely costly and difficult to produce them in sufficient numbers and with sufficient suppressive capacity to significantly impact the alloimmune response. Here we have used our established non-human primate model to demonstrate that significant Treg expansion (up to 600-fold in 21 days) can be maintained, and suppressive capacity enhanced by exposing Treg cultures to a short burst of sirolimus at the end of the culture period. Using a highly sensitive and specific in vitro CFSE-MLR assay we show that Tregs significantly inhibit allo-proliferation of multiple T cell subpopulations including both CD4+ and CD8+ T cells (3.2 and 2.7-fold inhibition of proliferation, respectively), as well as their CD28+CD95+ and CD28-CD95+ subpopulations (2.2 and 2.1 and 1.9 and 2.7-fold inhibition of CD4+ and CD8+ subpopulation proliferation, respectively). Tregs were able to combine in vitro with the newly FDA-approved CTLA4-Ig analog belatacept to enhance the inhibition of alloproliferation that occurred with either agent alone (4.8-fold inhibition of CD8 T cell proliferation with Tregs + belatacept, compared to 3.0-fold or 1.9-fold inhibition of CD8 T cell proliferation with Tregs or belatacept alone, respectively). Importantly, we have found that the suppressive activity of ex-vivo expanded Tregs could be further enhanced by pulsing with sirolimus. Thus, while long-term culture of Tregs in the presence of sirolimus (1–1000 nM) profoundly inhibited Treg expansion (50–800 fold inhibition of expansion when cultured in the presence of 1–1000 nM sirolimus), a 48 hour pulse of sirolimus (100 nM) on days 20–21 of culture completely preserved Treg yields while doubling their suppressive function against CD8 proliferation when compared to unpulsed Tregs, p<0.01) A mechanistic evaluation of the increase potency observed with sirolimus pulsed Tregs (SPTs) has revealed several key differences that distinguish these cells from the less-potent unpulsed Tregs: SPTs were found to undergo fewer rounds of proliferation in an MLR when compared with unpulsed Tregs (14% proliferation in SPTs versus 37% proliferation in un-pulsed Tregs, p= 0.015), suggesting that the suppressive capability of Tregs may be inversely related to their proliferative capacity. SPTs were also shown to have significantly increased expression of CD25 (p=0.04) and total CTLA4 (p= 0.009) compared to unpulsed Tregs, implicating signaling through both of these molecules in their enhanced function. Our results suggest that the creation of SPTs may provide a novel avenue by which to achieve enhanced Treg-based suppression of alloimmunity, in a manner that is amenable to large-scale ex-vivo expansion and to combinatorial therapy with novel, costimulation-blockade-based immunosuppression strategies. Disclosures: No relevant conflicts of interest to declare.


PLoS ONE ◽  
2015 ◽  
Vol 10 (12) ◽  
pp. e0144549 ◽  
Author(s):  
Young Ah Lee ◽  
Hang-Rae Kim ◽  
Jeong Seon Lee ◽  
Hae Woon Jung ◽  
Hwa Young Kim ◽  
...  

2005 ◽  
Vol 201 (11) ◽  
pp. 1793-1803 ◽  
Author(s):  
Claudia R. Ruprecht ◽  
Marco Gattorno ◽  
Francesca Ferlito ◽  
Andrea Gregorio ◽  
Alberto Martini ◽  
...  

A better understanding of the role of CD4+CD25+ regulatory T cells in disease pathogenesis should follow from the discovery of reliable markers capable of discriminating regulatory from activated T cells. We report that the CD4+CD25+ population in synovial fluid of juvenile idiopathic arthritis (JIA) patients comprises both regulatory and effector T cells that can be distinguished by expression of CD27. CD4+CD25+CD27+ cells expressed high amounts of FoxP3 (43% of them being FoxP3+), did not produce interleukin (IL)-2, interferon-γ, or tumor necrosis factor, and suppressed T cell proliferation in vitro, being, on a per cell basis, fourfold more potent than the corresponding peripheral blood population. In contrast, CD4+CD25+CD27− cells expressed low amounts of FoxP3, produced effector cytokines and did not suppress T cell proliferation. After in vitro activation and expansion, regulatory but not conventional T cells maintained high expression of CD27. IL-7 and IL-15 were found to be present in synovial fluid of JIA patients and, when added in vitro, abrogated the suppressive activity of regulatory T cells. Together, these results demonstrate that, when used in conjunction with CD25, CD27 is a useful marker to distinguish regulatory from effector T cells in inflamed tissues and suggest that at these sites IL-7 and IL-15 may interfere with regulatory T cell function.


2004 ◽  
Vol 174 (1) ◽  
pp. 164-173 ◽  
Author(s):  
Hideaki Sugiyama ◽  
Rolland Gyulai ◽  
Eiko Toichi ◽  
Edina Garaczi ◽  
Shinji Shimada ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2072-2072
Author(s):  
Christian P Pallasch ◽  
Susanne Ulbrich ◽  
Reinhild Brinker ◽  
Robert A Uger ◽  
Michael Hallek ◽  
...  

Abstract Suppression of patients’ T-cells is a key event in CLL pathogenesis and was demonstrated to be mediated by direct cell-cell contact of malignant CLL cells with T-cells. CD200 plays a critical role in regulating the immune system and has been shown to be up-regulated on the surface of different tumors including CLL. In this study we addressed the effects of CD200 over-expression on CLL cells on autologous T cells in a mixed lymphocyte reaction system. We used native and CD40 ligand (CD40L)- stimulated CLL cells as antigen-presenting cells (APCs) to expand autologous T cells of 14 patients. T-cell proliferation was analyzed over 3 weeks of in vitro culture. A functional anti- CD200 antibody (1B9) was added to reveal CD200-mediated immunosuppression in the autologous system. Expansion of patient T-cells was assessed by flow cytometry including intracellular staining of FOXP3. Specificity towards CLL-specific antigens was monitored applying fibromodulin derived peptides for detection of specific T-cells by ELISPOT analysis. T-cell proliferation over 3 weeks of in vitro culture was significantly enhanced compared to control cells when using CD40L-stimulated APCs and an anti-CD200 antibody (p=0.0004). CD200 blockade was further shown to stimulate antigen-specific T-cell responses towards the F2 and F4 peptides of the CLL-associated antigen fibromodulin (p=0.04). Finally, the number of CD4+/CD25high/FOXP3+ T cells (Treg) was significantly decreased in CD200 treated mixed lymphocyte reaction (p=0.04). In summary, CD200 blockade may provide therapeutic benefits in CLL by enhancing T-cell expansion, augmenting an antigen-specific T cell response with suppression of regulatory T cells. CD200 seems to be an important immunosuppressive molecule in CLL: by CD200 blockade immune suppression can be overcome by altering tolerance to tumor antigens and deregulation of regulatory T cells. This combination of an immune induction paralleled by a disruption of immunosuppressive factors makes anti-CD200 mAb a powerful tool for future treatment of CLL, possibly in combination with other B cell cytotoxic or immunostimulatory approaches.


2009 ◽  
Vol 131 (1) ◽  
pp. 109-118 ◽  
Author(s):  
Li-Song Shen ◽  
Jian Wang ◽  
Ding-Feng Shen ◽  
Xiang-Liang Yuan ◽  
Ping Dong ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document