scholarly journals Interleukin-5 (IL-5) Therapy Prevents Allograft Rejection by Promoting CD4+CD25+ Ts2 Regulatory Cells That Are Antigen-Specific and Express IL-5 Receptor

2021 ◽  
Vol 12 ◽  
Author(s):  
Bruce M. Hall ◽  
Rachael M. Hall ◽  
Giang T. Tran ◽  
Catherine M. Robinson ◽  
Paul L. Wilcox ◽  
...  

CD4+CD25+Foxp3+T cell population is heterogenous and contains three major sub-groups. First, thymus derived T regulatory cells (tTreg) that are naïve/resting. Second, activated/memory Treg that are produced by activation of tTreg by antigen and cytokines. Third, effector lineage CD4+CD25+T cells generated from CD4+CD25- T cells’ activation by antigen to transiently express CD25 and Foxp3. We have shown that freshly isolated CD4+CD25+T cells are activated by specific alloantigen and IL-4, not IL-2, to Ts2 cells that express the IL-5 receptor alpha. Ts2 cells are more potent than naïve/resting tTreg in suppressing specific alloimmunity. Here, we showed rIL-5 promoted further activation of Ts2 cells to Th2-like Treg, that expressed foxp3, irf4, gata3 and il5. In vivo, we studied the effects of rIL-5 treatment on Lewis heart allograft survival in F344 rats. Host CD4+CD25+T cells were assessed by FACS, in mixed lymphocyte culture and by RT-PCR to examine mRNA of Ts2 or Th2-like Treg markers. rIL-5 treatment given 7 days after transplantation reduced the severity of rejection and all grafts survived ≥60d whereas sham treated rats fully rejected by day 31 (p<0.01). Treatment with anti-CD25 or anti-IL-4 monoclonal antibody abolished the benefits of treatment with rIL-5 and accelerated rejection. After 10d treatment with rIL-5, hosts’ CD4+CD25+ cells expressed more Il5ra and responded to specific donor Lewis but not self. Enriched CD4+CD25+ cells from rIL-5 treated rats with allografts surviving >60 days proliferated to specific donor only when rIL-5 was present and did not proliferate to self or third party. These cells had more mRNA for molecules expressed by Th2-like Treg including Irf4, gata3 and Il5. These findings were consistent with IL-5 treatment preventing rejection by activation of Ts2 cells and Th2-like Treg.

1983 ◽  
Vol 157 (1) ◽  
pp. 141-154 ◽  
Author(s):  
P J Fink ◽  
I L Weissman ◽  
M J Bevan

To detect a strong cytotoxic T lymphocyte (CTL) response to minor histocompatibility (H) antigens in a 5-d mixed lymphocyte culture, it is necessary to use a responder that has been primed in vivo with antigen-bearing cells. It has previously been shown that minor-H-specific CTL can be primed in vivo both directly by foreign spleen cells and by presentation of foreign minor H antigens on host antigen-presenting cells. This latter route is evident in the phenomenon of cross-priming, in which H-2 heterozygous (A x B)F1 mice injected 2 wk previously with minor H-different H-2A (A') spleen cells generate both H-2A- and H-2B-restricted minor-H-specific CTL. In a study of the kinetics of direct- vs. cross-priming to minors in F1 mice, we have found that minor H-different T cells actually suppress the induction of virgin CTL capable of recognizing them. CTL activity measured from F1 mice 3-6 d after injection with viable A' spleen cells is largely H-2B restricted. The H-2A-restricted response recovers such that roughly equal A- and B-restricted activity is detected in mice as early as 8-10 d postinjection. This temporary hyporeactivity does not result from generalized immunosuppression--it is specific for those CTL that recognize the foreign minor H antigen in the context of the H-2 antigens on the injected spleen cells. The injected spleen cells that mediate this suppression are radiosensitive T cells; Lyt-2+ T cells are highly efficient at suppressing the induction of CTL in vivo. No graft vs. host reaction by the injected T cells appears to be required, as suppression of direct primed CTL can be mediated by spleen cells that are wholly tolerant of both host H-2 and minor H antigens. Suppression cannot be demonstrated by in vitro mixing experiments. Several possible mechanisms for haplotype-specific suppression are discussed, including inactivation of responding CTL by veto cells and in vivo sequestration of responding CTL by the injected spleen cells.


2017 ◽  
Vol 37 (suppl_1) ◽  
Author(s):  
Dennis Wolf ◽  
Teresa Gerhardt ◽  
Nathaly Anto Michel ◽  
Bjarke Hansen ◽  
Alessandro Sette ◽  
...  

Background: In atherosclerosis, CD4 + T helper cells recognize auto-antigens including ApoB, the main protein in low-density lipoprotein (LDL). However, atherosclerosis-specific, auto-reactive CD4 + T cells have not been detected in vivo , and their function is unknown. Methods and Results: We have previously identified peptides derived from mouse ApoB that bind with high affinity to the MHC class II molecule of C57BL/6 mice (I-A b ). We designed and validated a new multimer of a recombinant MHC-II molecule fused to one ApoB auto-epitopes, P6 (TGAYSNASSTESASY, P6:I-A b ), that enabled detection of low-affinity, P6-reactive CD4 + T cells. Using this P6:I-A b multimer, we identified ApoB-reactive CD4 + T cells in healthy, young C57BL/6 mice that were predominately differentiated T-regulatory cells (T regs ) and expressed IL-10, a known atheroprotective cytokine. This population was detectable in lymph nodes and already showed a memory phenotype in young animals without atherosclerosis. In Apoe -/- mice, adoptively transferred ApoB P6-specific T regs accumulated in the aorta and draining lymph nodes and gave rise to pathogenic T H 1 and T H 17 cells. This phenotypic switch was caused by enhanced plasticity of antigen-specific T regs as evidenced by multiple clusters of intermediate T reg -T eff phenotypes in single cell RNA sequencing of 4485 antigen-specific CD4 + T cells. In the plaque, many T cells were ex-T regs as identified by a FoxP3 lineage tracker mouse, suggesting that atherosclerosis-specific CD4 + T cells lost their regulatory capacity. Vaccination with P6 maintained a protective phenotype in antigen-specific T regs and protected from atherosclerosis. In humans, ApoB-specific CD4 + T cells from atherosclerotic patients showed the same cytokine patterns found in mouse CD4 + T cells, suggesting that autoimmunity to ApoB is protective first, but later gives rise to a pathogenic CD4 + T cell response that aggravates atherosclerosis. Conclusion: Protective T-regulatory cells recognizing peptide antigens of ApoB exist in naïve mice, protect against atherosclerosis, but convert into pathogenic T H 1 and -17 cells during the natural course of disease in mice and humans. These results call for immunomodulatory therapies to maintain protective autoimmunity.


1976 ◽  
Vol 144 (3) ◽  
pp. 617-626 ◽  
Author(s):  
J Sprent ◽  
H von Boehmer

Helper T cells were obtained by injecting heavily irradiated semiallogeneic mice with lymph node cells from H-2-incompatible parental strain mice primed with sheep erythrocytes (SRC) 2 mo before. Thoracic duct lymphocytes collected from the recipents 18-40 h later (nearly all of which were theta-positive and of donor origin) were totally and specifically unresponsive against host-type determinants in mixed-lymphocyte culture. The filtered cells were transferred to irradiated semiallogeneic mice together with SRC and anti-theta-serum-treated (B) cells from SRC-primed syngeneic, semiallogeneic, or allogeneic mice. When antibody-forming cells were measured in the spleen 5-9 days later, effective IgM and IgG collaborative responses were observed with both syngeneic and semiallogeneic B cells but not with allogeneic B cells. No evidence was found that the failure to obtain collaboration with the allogeneic B cells was due to inhibition of the B cells by the T cells or vice versa.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1307-1307
Author(s):  
Robert B. Levy ◽  
Angela Jones

Abstract CD4 regulatory T (Treg) cells have shown promise in the transplantation mileu including the ability to inhibit the development of graft vs host disease (GVHD) following allogeneic hematopoietic stem cell transplants (HCT). The antigen specificity of the Treg population(s) involved is not yet clear nor is the role of their activation following transplant. We are interested in determining the requirement for recognition of host MHC antigens following infusion of CD4+CD25+ T cells in an experimental model of GVHD. To clearly distinguish the requirements of regulatory vs GVH reactive cells, a model of CD8 T cell mediated GVHD was developed using highly purified BALB/c (H2d) donor CD8+ T cells (Miltenyi column, 95-98%). CD8 T cells were transplanted together with T cell depleted (TCD) BALB/c BMC into 12.0 GY (6.0 Gy split dose) TBI conditioned C57BL/6 (B6, H2b) recipients. To support development of GVHD by these cells, resistance was inhibited by treatment of recipients with anti-NK1.1mab (PK136) at Days -1, 0 and +7. BALB/c CD8+ T cells at doses of 5.0x106 but not 2.5x106 induced weight loss and some lethality in B6 recipients. 5x106 CD8+ T cells were then transplanted into B6-MHC class II−/ − recipients. GVHD symptoms including weight loss and lethality were readily apparent in these mice post-transplant. Interestingly, GVHD was consistently more severe with respect to the induction of weight loss and lethality in MHC Class II−/ − vs B6-wt recipients. Highly enriched BALB/c CD4+CD25+ T cells (> 95%) were produced from spleen and lymph node cells following negative (B-cells, CD8 and NK) and positive (CD25) selection using Miltenyi magnetic bead columns. Co-transplant of 1x106 CD4+CD25+ T cells together with BALB/c CD8+ T cells into B6 recipients inhibited GVHD as assessed by the absence of weight loss and lethality compared to B6 recipients of CD8+ T cells alone. In contrast, BALB/c CD4+CD25+ T cells failed to protect B6-MHC class II−/ − recipients from severe CD8+ T cell mediated GVHD. These findings demonstrate that donor CD4+ T regulatory cells can suppress GVHD inducing CD8+ T cells after the former recognize host class II alloantigen following transplant. We hypothesize that activated CD4+CD25+ T regulatory cells inhibit GVH reactive T cells at the host APC interface. Future studies in this model can be designed to examine ex-vivo activated and expanded CD4+CD25+ T regulatory populations. Transplant of such cells will enable us to address questions regarding the importance of in vivo recognition of host class II in the regulation of GVHD by these cells.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2342-2342
Author(s):  
Rick Kapur ◽  
Michael Kim ◽  
Shanjee Shanmugabhavananthan ◽  
Edwin R. Speck ◽  
Rukhsana Aslam ◽  
...  

Abstract Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related fatalities and is characterized by acute respiratory distress following transfusion of blood products. Frequently, donor antibodies present in the transfused blood product are involved, such as anti-human leukocyte antigen (HLA) antibodies or anti-human neutrophil antigen (HNA) antibodies. Several animal models of TRALI have contributed to understanding the pathogenesis which, however, is still incompletely understood. Several cell types have also been suggested to be involved in antibody-mediated TRALI, including neutrophils, endothelial cells and monocytes. Most of the animal models are based on a two-hit paradigm, where the first hit is based on "patient predisposition", such as sepsis or chronic alcohol abuse, while the second hit is delivered in the form of "transfusion factors", such as antibodies present in the transfused blood product. Although most studies have focused on factors contributing to the development of antibody-mediated TRALI, the factors and mechanisms in place to protect against antibody-mediated TRALI have been underexplored. Adoptive transfer of lymphocytes into recipient severe combined immunodeficient (SCID) mice, in which the well-established TRALI inducing anti-MHC class I antibody clone 34-1-2s was injected, was previously shown to rescue TRALI induction by 34-1-2s. Here we describe, using a murine BALB/c antibody-mediated TRALI model based on injection of 34-1-2s, that CD4 T cells, and more specifically, CD4+CD25+Foxp3+ T regulatory cells (Tregs), are responsible for protection against murine antibody-mediated TRALI. Specific in vivo depletion of CD4+ T cells, or targeted in vivo depletion of Tregs, resulted in severe lung damage after 34-1-2s infusion, as determined by increased lung wet-to-dry ratios (a measure for pulmonary edema), generally greater than 5, indicative of severe pulmonary edema. This was accompanied by significant hypothermia, increased values of the neutrophil chemoattractant macrophage inflammatory protein 2 (MIP-2: equivalent of human IL-8), and increased pulmonary neutrophil accumulation, all compared to control groups. In contrast, systematic in vivo depletion of CD8+ T cells, B cells or monocytes, did not result in significant lung damage. Co-depletion of CD4+ T cells together with monocytes rescued the TRALI induction by 34-1-2s, validating the pathogenic role of monocytes in murine antibody-mediated TRALI induction. Based on MIP-2 values and in vitro studies, we suggest that Tregs suppress monocytes in order to prevent antibody-mediated TRALI. Overall, a novel first hit in TRALI induction could be identified in conditions that cause a decrease in Treg number or function, which could also explain the increased risk for human TRALI in cases of chronic alcohol abuse. In addition, therapies aimed at restoring Treg numbers or function may prove to be a novel therapeutic approach in antibody-mediated TRALI. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1229-1229
Author(s):  
Benny J. Chen ◽  
Xiuyu Cui ◽  
Nelson J. Chao

Abstract We and others have recently demonstrated that memory T cells do not induce graft-versus-host disease in several different animal models. To test whether the same concept applies to humans, we compared the ability of memory T cells to respond to alloantigens with that of naive as well as bulk T cells. Purified T cells were first obtained from peripheral blood from healthy donors and then separated into memory and naive T cell subsets based on the expression of CD45RA (memory: CD45RA−, naive: CD45RA+). Memory T cells were subsequently tested for their ability to respond to alloantigens using proliferation and cytotoxicity assays in comparison with naive and bulk T cells. Proliferation assay was performed using 1.25x105 responder cells and 5x105 irradiated stimulator cells per well in 96-well flat-bottom plate. Cytotoxicity was measured by the standard 4-hour Cr-51 release assay after 5-day mixed lymphocyte culture. In contrast to the mouse data, memory T cells proliferated equally well as naive and bulk T cells did in mixed lymphocyte culture (Figure A). However, these same memory T cells failed to kill the allogeneic targets despite the vigorous proliferative responses against the same alloantigens (Figure B). These data demonstrated that human memory T cells proliferate but do not elicit cytotoxicity in response to alloantigens, suggesting that human memory T cells may not contain true alloantigen-specific T cells if they have never exposed to those alloantigens before and may not cause graft-versus-host disease upon in vivo transfer. These observations warrant the further testing of human memory T cells in clinically more relevant models and in vivo for their ability to induce graft-versus-host disease. Figure Figure


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3813-3813
Author(s):  
Jessica Lauren Heinrichs ◽  
Jun Li ◽  
Jianing Fu ◽  
Xue-Zhong Yu

Abstract Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a therapeutic option for hematologic malignances. Donor T effector cells (Teffs), recognizing mismatched major and minor histocompatibility antigens, are both beneficial and detrimental, as they mediate anti-tumor response (GVL) and graft-versus-host disease (GVHD), the latter of which remains a significant complication arising after allo-HSCT. Adoptive cellular therapy using polyclonal natural T regulatory (nTreg) has been shown to be a promising strategy in the prevention and treatment of GVHD. However, clinical application of nTreg therapy requires high doses and expanded nTregs have sparse availability, limited expansion capacity, and potential contamination with Teffs. Induced T regulatory cells (iTregs) could solve these limitations given their robust proliferative potential and increased availability. Moreover, iTregs can be easily educated with alloantigen prior to clinical infusion, increasing their selectivity and potency, which may result in a more targeted and effective cellular therapy compared to polyclonal nTregs. In the current work, we tested the hypothesis that alloreactive iTregs can be readily generated and possess high potency to suppress GVHD with better selectivity. By stimulating CD4+CD25- T cells with allogeneic dendritic cells (DCs) in the presence of IL-2, TGF-β, and retinoic acid, high percentages of CD25+Foxp3+ iTregs (60-80%) were induced in 5-day culture. These alloreactive iTregs were highly suppressive in T-cell responses to the same allogeneic stimulation, and were 16-32 fold more suppressive than those polyclonal iTregs generated by αCD3/αCD28-stimulation in vitro. Using a well-defined, fully MHC-mismatched B6 (H2b) to Balb/c (H2d) murine model of allogeneic BMT, we demonstrated that co-transfer of donor iTregs (H2b) generated with recipient DCs (H2d) significantly suppressed GVHD mortality, whereas the donor iTregs (H2b) generated with a third-party DCs (H2k) were significantly less effective. In mechanistic studies, we found that iTregs reactive to recipient alloantigens (H2d) expanded more dramatically and were significantly more stable in terms of maintaining Foxp3 expression as compared to the iTregs reactive to third-party alloantigens (H2k) in H2drecipients. As a consequence, recipient-reactive iTregs were significantly more effective than third-party reactive iTregs in inhibiting activation, expansion, and migration of Teffs into GVHD target organs. Although alloreactive CD4 iTregs were effective in alleviating GVHD, high dose and multiple infusions were necessary to prevent GVHD lethality for the majority of recipients. Published studies indicate that CD8 iTregs are generated in allogeneic recipients after allo-BMT in vivo and also contribute to the regulation of GVHD development. Aiming to improve Treg efficacy, we hypothesized that alloreactive CD8 iTregs can be generated in vitro and used for singular or combinational cell therapy. To this end, we established a protocol to induce alloreactive CD8 iTregs, and evaluated their suppressive activity. While these CD8 iTregs were potently suppressive of alloresponse in vitro, they were only partially protective in vivo in attenuating GVHD. Given we also observed that CD8 iTregs were superior to CD4 iTregs in the suppression of CD8 Teff responses to alloantigens in vitro, we further hypothesized that a combinational therapy with CD4 and CD8 iTregs would be more effective in the prevention of GVHD than either alone. Indeed we found the combined CD4 and CD8 iTreg therapy to be significantly more potent in attenuating GVHD and reducing clinical score than either single therapy alone. In conclusion, alloreactive CD4 iTregs are significantly more effective than polyclonal counterparts in suppressing alloresponse in vitro and preventing GVHD in vivo. These alloreactive CD4 iTregs prevent GVHD by inhibiting CD4+ Teff expansion and migration through an antigen-dependent manner. For the first time, to our knowledge, we showed that alloreactive CD8 iTregs were highly potent in suppressing alloresponse in vitro and moderately potent in preventing GVHD in vivo. Furthermore, we provided evidence indicating that perfecting adoptive iTreg therapy may require a combination of CD4 and CD8 iTregs to achieve maximum efficacy in the prevention of GVHD. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 113 (18) ◽  
pp. 4458-4467 ◽  
Author(s):  
Asha B. Pillai ◽  
Tracy I. George ◽  
Suparna Dutt ◽  
Samuel Strober

Abstract Although CD4+CD25+ T cells (T regulatory cells [Tregs]) and natural killer T cells (NKT cells) each protect against graft-versus-host disease (GVHD), interactions between these 2 regulatory cell populations after allogeneic bone marrow transplantation (BMT) have not been studied. We show that host NKT cells can induce an in vivo expansion of donor Tregs that prevents lethal GVHD in mice after conditioning with fractionated lymphoid irradiation (TLI) and anti–T-cell antibodies, a regimen that models human GVHD-protective nonmyeloablative protocols using TLI and antithymocyte globulin (ATG), followed by allogeneic hematopoietic cell transplantation (HCT). GVHD protection was lost in NKT-cell–deficient Jα18−/− hosts and interleukin-4 (IL-4)−/− hosts, or when the donor transplant was Treg depleted. Add-back of donor Tregs or wild-type host NKT cells restored GVHD protection. Donor Treg proliferation was lost in IL-4−/− hosts or when IL-4−/− mice were used as the source of NKT cells for adoptive transfer, indicating that host NKT cell augmentation of donor Treg proliferation after TLI/antithymocyte serum is IL-4 dependent. Our results demonstrate that host NKT cells and donor Tregs can act synergistically after BMT, and provide a mechanism by which strategies designed to preserve host regulatory cells can augment in vivo donor Treg expansion to regulate GVHD after allogeneic HCT.


Sign in / Sign up

Export Citation Format

Share Document