scholarly journals The complement inhibitory protein DAF (CD55) suppresses T cell immunity in vivo

2005 ◽  
Vol 201 (4) ◽  
pp. 567-577 ◽  
Author(s):  
Jianuo Liu ◽  
Takashi Miwa ◽  
Brendan Hilliard ◽  
Youhai Chen ◽  
John D. Lambris ◽  
...  

Decay-accelerating factor ([DAF] CD55) is a glycosylphosphatidylinositol-anchored membrane inhibitor of complement with broad clinical relevance. Here, we establish an additional and unexpected role for DAF in the suppression of adaptive immune responses in vivo. In both C57BL/6 and BALB/c mice, deficiency of the Daf1 gene, which encodes the murine homologue of human DAF, significantly enhanced T cell responses to active immunization. This phenotype was characterized by hypersecretion of interferon (IFN)-γ and interleukin (IL)-2, as well as down-regulation of the inhibitory cytokine IL-10 during antigen restimulation of lymphocytes in vitro. Compared with wild-type mice, Daf1−/− mice also displayed markedly exacerbated disease progression and pathology in a T cell–dependent experimental autoimmune encephalomyelitis (EAE) model. However, disabling the complement system in Daf1−/− mice normalized T cell secretion of IFN-γ and IL-2 and attenuated disease severity in the EAE model. These findings establish a critical link between complement and T cell immunity and have implications for the role of DAF and complement in organ transplantation, tumor evasion, and vaccine development.

1999 ◽  
Vol 190 (7) ◽  
pp. 1033-1038 ◽  
Author(s):  
Jan Paul Medema ◽  
Joan de Jong ◽  
Thorbald van Hall ◽  
Cornelis J.M. Melief ◽  
Rienk Offringa

The antiapoptotic protein cellular FLICE (Fas-associated death domain–like IL-1β–converting enzyme) inhibitory protein (cFLIP) protects cells from CD95(APO-1/Fas)-induced apoptosis in vitro and was found to be overexpressed in human melanomas. However, cytotoxic T cell–induced apoptosis, which is critically involved in tumor control in vivo, is not inhibited by cFLIP in vitro, as only CD95- and not perforin-dependent lysis is affected. This calls into question whether cFLIP is sufficient to allow escape from T cell–dependent immunity. Using two murine tumors, we directly demonstrate that cFLIP does result in escape from T cell immunity in vivo. Moreover, tumor cells are selected in vivo for elevated cFLIP expression. Therefore, our data indicate that CD95-dependent apoptosis constitutes a more prominent mechanism for tumor clearance than has so far been anticipated and that blockade of this pathway can result in tumor escape even when the perforin pathway is operational.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3240-3240
Author(s):  
Hannah Cullup ◽  
John Wilson ◽  
Alison M. Rice ◽  
Anne M. Dickinson ◽  
David J. Munster ◽  
...  

Abstract AGVHD is a life threatening complication of allogeneic HSCT, initiated by host and donor DC stimulation of donor T lymphocytes. Current AGVHD prophylaxis targets T lymphocytes, compromising anti-viral and therapeutic anti-leukemic responses. The presence of activated blood DC predicts for clinical AGVHD and we predict that a new strategy targeting therapy to activated DC should prevent alloimmune induction of AGVHD but preserve protective T cell immune responses. CD83 is a cell surface molecule expressed by activated DC. Having shown that a rabbit polyclonal antibody to human CD83 (RA83) depletes activated DC and suppresses alloimmune reactions in vitro, we tested the effect of RA83 treatment on T cell immunity in vitro and its ability to prevent human PBMC induced xenogeneic AGVHD in vivo. The effect of RA83 treatment on T cell numbers, proliferation and cytokine secretion in allogeneic MLR was compared with appropriate controls, including Campath-1H. Allogeneic responses in vitro were mirrored in a DC dependent in vivo model of xenogeneic AGVHD, in which 50×106 human PBMC were injected into an irradiated SCID mouse. Human cytokine levels were measured in MLR tissue culture supernatant (TCSN) and mouse serum at the time of sacrifice. Cytotoxic T cell responses to viral antigens (CMV and FMP) were analysed by specific pentamer analysis and Cr51 release assays, prior to and following HLA restricted peptide antigen specific expansion. RA83 was shown to have NK-mediated ADCC capacity. Cellular proliferation in the allogeneic MLR was reduced by both RA83 and Campath-1H treatment (p= 0.004 and 0.01 respectively vs controls) and both antibodies improved mouse survival in the human xenogenic AGVHD model (RA83: 93%, Campath-1H: 100%, p<0.0001). IFN-g was significantly reduced in the TCSN from MLR treated with RA83 (p=0.0391) and in sera taken from RA83 (p=0.0002) and Campath-1H (p=0.0051) treated mice. Serum IL-4 levels were maintained in RA83 and Campath-1H treated mice. The serum levels of IL-5, IL-8 and TNF in mice treated with RA83 were markedly reduced compared to controls (p=0.0256, 0.0025, 0.025, respectively), and the reductions were similar to those seen in Campath-1H treated mice. Similar numbers of T cells were recovered from RA83 treated and control MLR, and both CMV and FMP specific CD8+ T cells were retained. These cells were readily expanded by peptide pulsing and autologous restimulation and had specific cytotoxic activity comparable to control cultures (see figure). In contrast, Campath-1H treatment removed specific anti-viral responses (vs controls: CMV: p<0.00001 and FMP: p=0.0051). Specific antibody to CD83 depletes activated DC in vitro and prevents xenogeneic human DC dependent AGVHD in vivo. This was accompanied by a Th1 to Th2 skewing of the cytokine response. RA83, but not Campath-1H treatment retained normal numbers of T cells and maintained normal cytotoxic responses to common post-transplant viral infections. Depletion of activated DC may be an effective means of AGVHD control, which maintains T cell immunity to life threatening infections and potentially anti-leukaemia responses. RA83 treatment of allo-MLR preserves T cell anti-CMV immunity RA83 treatment of allo-MLR preserves T cell anti-CMV immunity


Author(s):  
Luo Li ◽  
Qian Chen ◽  
Xiaojian Han ◽  
Meiying Shen ◽  
Chao Hu ◽  
...  

A better understanding of the role of T cells in the immune response to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is helpful not only for vaccine development but also for the treatment of COVID-19 patients. In this study, we determined the existence of SARS-CoV-2-specific T cells in the blood of COVID-19 convalescents. Meanwhile, the specific T cell response in the non-RBD region was stronger than in the RBD region. We also found that SARS-CoV-2 S-specific reactive CD4+ T cells exhibited higher frequency than CD8+ T cells in recovered COVID-19 patients, with greater number of corresponding epitopes presented. Importantly, we isolated the SARS-CoV-2-specific CD4+ T cell receptors (TCRs) and inserted the TCRs into allogenic CD4+ T cells. These TCR-T cells can be activated by SARS-CoV-2 spike peptide and produce IFN-γ in vitro. These results might provide valuable information for the development of vaccines and new therapies against COVID-19.


Blood ◽  
2002 ◽  
Vol 99 (6) ◽  
pp. 2146-2153 ◽  
Author(s):  
James L. LaBelle ◽  
Carrie A. Hanke ◽  
Bruce R. Blazar ◽  
Robert L. Truitt

Abstract B7 molecules provide important costimulatory signals to T cells, and B7 genes have been introduced into B7-negative tumor cells to enhance their immunogenicity. However, the role of B7 molecules in inducing tumor immunity is controversial because of conflicting results and reports of differential signaling through the B7 molecules and their ligands CD28 and CTLA-4. In this study, we compared the effect of B7-1 (CD80) and B7-2 (CD86) on the induction of T-cell immunity to C1498, a murine myelogenous leukemia. When cultured with exogenous cytokines in vitro, C1498/B7-1 and C1498/B7-2 induced syngeneic CD8+ T cells to kill parental C1498. In vivo, C1498/B7-1 grew progressively after subcutaneous injection, whereas C1498/B7-2 completely regressed after transient growth in naive mice. Spontaneous rejection of C1498/B7-2 resulted in immunity to challenge doses of C1498 and C1498/B7-1. Antibody-depletion studies in vivo showed that CD8+ T cells rejected C1498/B7-2, whereas only natural killer cells affected the growth of C1498/B7-1. Two approaches were used to determine whether preferential interaction of B7-1 with CTLA-4 contributed to the failure of C1498/B7-1 to activate CD8+ T cells in vivo. First, CTLA-4 specific monoclonal antibody was used to block B7-1–CTLA-4 interaction. Second, CTLA-4 deletional mutant (−/−) bone marrow chimeras were used as tumor hosts. In both systems, there was a significant increase in the rate of rejection of C1498/B7-1 tumors. Resistance to C1498/B7-1 in CTLA-4−/− hosts was mediated by CD8+ T cells. Blocking or deletion of CTLA-4 did not affect the growth of parental C1498, indicating that B7-1 was important for the induction of CD8+ T-cell immunity in the absence of CTLA-4.


2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii472-iii472
Author(s):  
Mubeen Mosaheb ◽  
Daniel Landi ◽  
Elena Dobrikova ◽  
Michael Brown ◽  
Yuanfan Yang ◽  
...  

Abstract BACKGROUND H3 K27M-mutant diffuse midline glioma (DMG) is invariably lethal. Viruses naturally engage innate immunity, induce antigen presentation, and mediate CD8 T cell priming against foreign antigens. Polioviruses, in particular, are uniquely tropic for dendritic cells (DC) and potently activate DC, inducing Th1-dominant cytokine profiles, CD8 T cell immunity, and enhanced epitope presentation. Thus, poliovirus is ideally suited for vectored delivery of signature tumor neoantigens, e.g. the H3 K27M feature of DMG. However, poliovirus vector design is inherently limited by genetic instability and the underlying neuropathogenicity of poliovirus. METHODS We created a genetically stable, polio:rhinovirus chimera vector devoid of neuropathogenicity and modified for stable expression of the HLA-A2 restricted H3.3 K27M antigen (RIPO (H3.3)). RESULTS RIPO(H3.3) infects, activates, and induces H3.3K27M antigen presentation in DCs in vitro. Given intramuscularly in vivo, RIPO(H3.3) recruits and activates DCs with Th1-dominant cytokine profiles, efficiently primes H3.3K27M-specific CD8 T cells, induces antigen-specific CD8 T cell migration to the tumor site, delays tumor growth, and enhances survival in murine tumor models. CONCLUSION This novel approach leverages the unique ability of polioviruses to activate DCs while simultaneously introducing the H3.3 K27M antigen. In this way, DCs are activated optimally in situ, while being simultaneously infected to express/present tumor antigen. RIPO(H3.3), given by intramuscular injection, will be evaluated in a clinical trial for children with H3 K27M-mutant diffuse midline glioma.


2011 ◽  
Vol 188 (2) ◽  
pp. 892-901 ◽  
Author(s):  
Xiang Xiao ◽  
Weihua Gong ◽  
Gulcin Demirci ◽  
Wentao Liu ◽  
Silvia Spoerl ◽  
...  

2017 ◽  
Vol 7 (1) ◽  
Author(s):  
Liangquan Zhu ◽  
Xinxin Zhao ◽  
Qing Yin ◽  
Xianyong Liu ◽  
Xiang Chen ◽  
...  

2017 ◽  
Vol 38 (8) ◽  
pp. 594-605 ◽  
Author(s):  
Noemia S. Lima ◽  
Morgane Rolland ◽  
Kayvon Modjarrad ◽  
Lydie Trautmann

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A737-A737
Author(s):  
Anna Cole ◽  
Guillermo Rangel RIvera ◽  
Aubrey Smith ◽  
Megan Wyatt ◽  
Brandon Ware ◽  
...  

BackgroundIL-21 enhances the anti-tumor capacity of adoptively transferred CD8+ T cells, while IL-2 and IL-15 impair T cell immunity by driving their expansion to a more differentiated status. Yet, these cytokines can act on many different immune cells. Given the potency of IL-21, we tested if this cytokine directly augments T cells or rather if it enhances other immune cells in the culture that indirectly improves T cell therapy.MethodsTo test this question, splenocytes from pmel-1 transgenic mice were used, as all CD8+ T cells express a transgenic TCR specific for tumor-antigen gp10025–33 overexpressed on melanoma. We then peptide activated naïve CD8+ T cells enriched or not from the spleen of pmel-1 mice and expanded them in the presence of IL-21 or IL-2 (10 ng/mL) for four days. Expanded pmel-1 from these various cultures were then restimulated with irradiated splenocytes pulsed with gp10025–33 and grown an additional seven days with IL-2 (10 ng/mL), irrespective of their initial cytokine condition. The in vitro memory phenotype, exhaustion profile, and cytokine secretion of these cultures were then assayed. Furthermore, mice bearing B16KVP melanoma tumors were infused with pmel-1 T cells expanded via these various approaches and compared for their relative capacity to engraft, persist, and regress tumor in vivo.ResultsInterestingly, we discovered that IL-21-treated T cells generated from bulk splenocytes are phenotypically and functionally distinct from IL-21-treated isolated T cells. Upon restimulation, IL-21-treated T cells from bulk splenocytes exhibited an exhausted phenotype that was like anergic IL-2-treated T cells. Moreover, few cells expressed CD62L but expressed heightened markers of suppression, including TIM3, PD-1, and EOMES. Moreover, they produced more effector molecules, including granzyme B and IFN-gamma. In vivo IL-21-treated T cells expanded from bulk splenocytes engrafted and persisted poorly, in turn mediating suboptimal regression of melanoma. Conversely, IL-21 dramatically bolstered the engraftment and antitumor activity of T cells only if they were first isolated from the spleen prior to their expansion and infusion into the animal.ConclusionsCollectively, our data shows that IL-21 may improve ACT therapy best when used directly on antitumor CD8+ T cells. Further studies will illuminate the mechanism behind this striking difference and determine whether other cell subsets reactive to IL-21 cause T cell dysfunction and/or reduced bioavailability. These findings are important for defining the best culture conditions in which to use IL-21 for ACT.AcknowledgementsWe would like to acknowledge Emory University, The Winship Cancer Institute, and the Pediatrics/Winship Flow Cytometry Core.Ethics ApprovalAll animal procedures were approved by the Institutional Animal Care and Use Committee of Emory University, protocol number 201900225.


Sign in / Sign up

Export Citation Format

Share Document