scholarly journals TIM-3 drives temporal differences in restimulation-induced cell death sensitivity in effector CD8+ T cells in conjunction with CEACAM1

2021 ◽  
Vol 12 (4) ◽  
Author(s):  
Camille M. Lake ◽  
Kelsey Voss ◽  
Bradly M. Bauman ◽  
Katherine Pohida ◽  
Timothy Jiang ◽  
...  

AbstractImmune homeostasis depends upon effective clearance of pathogens while simultaneously preventing autoimmunity and immunopathology in the host. Restimulation-induced cell death (RICD) is one such mechanism where by activated T cells receive subsequent antigenic stimulation, reach a critical signal threshold through the T cell receptor (TCR), and commit to apoptosis. Many details of this process remain unclear, including the role of co-stimulatory and co-inhibitory proteins that influence the TCR signaling cascade. Here we characterize the role of T cell immunoglobulin and mucin domain containing 3 (TIM-3) in RICD regulation. TIM-3 protected newly activated CD8+ effector T cells from premature RICD during clonal expansion. Surprisingly, however, we found that TIM-3 potentiated RICD in late-stage effector T cells. The presence of TIM-3 increased proximal TCR signaling and proapoptotic protein expression in late-stage effector T cells, with no consistent signaling effects noted in newly activated cells with or without TIM-3. To better explain these differences in TIM-3 function as T cells aged, we characterized the temporal pattern of TIM-3 expression in effector T cells. We found that TIM-3 was expressed on the surface of newly activated effector T cells, but remained largely intracellular in late-stage effector cells. Consistent with this, TIM-3 required a ligand to prevent early RICD, whereas ligand manipulation had no effects at later stages. Of the known TIM-3 ligands, carcinoembryonic antigen‐related cell adhesion molecule (CEACAM1) showed the greatest difference in surface expression over time and also protected newly activated cells from premature RICD, with no measurable effects in late-stage effectors. Indeed, CEACAM1 enabled TIM-3 surface expression on T cells, implying a co-dependency for these proteins in protecting expanding T cells from premature RICD. Our findings suggest that co-signaling proteins like TIM-3 and CEACAM1 can alter RICD sensitivity at different stages of the effector T cell response, with important implications for checkpoint blockade therapy.

2009 ◽  
Vol 29 (21) ◽  
pp. 5952-5962 ◽  
Author(s):  
Caryll Waugh ◽  
Linda Sinclair ◽  
David Finlay ◽  
Jose R. Bayascas ◽  
Doreen Cantrell

ABSTRACT The present study explored the consequences of phosphoinositide (3,4,5)-triphosphate [PI(3,4,5)P3] binding to the pleckstrin homology (PH) domain of the serine/threonine kinase 3-phosphoinositide-dependent kinase 1 (PDK1). The salient finding is that PDK1 directly transduces the PI(3,4,5)P3 signaling that determines T-cell trafficking programs but not T-cell growth and proliferation. The integrity of the PDK1 PH domain thus is not required for PDK1 catalytic activity or to support cell survival and the proliferation of thymic and peripheral T cells. However, a PDK1 mutant that cannot bind PI(3,4,5)P3 cannot trigger the signals that terminate the expression of the transcription factor KLF2 in activated T cells and cannot switch the chemokine and adhesion receptor profile of naïve T cells to the profile of effector T cells. The PDK1 PH domain also is required for the maximal activation of Akt/protein kinase B (PKB) and for the maximal phosphorylation and inactivation of Foxo family transcription factors in T cells. PI(3,4,5)P3 binding to PDK1 and the strength of PKB activity thus can dictate the nature of the T-cell response. Low levels of PKB activity can be sufficient for T-cell proliferation but insufficient to initiate the migratory program of effector T cells.


Blood ◽  
2007 ◽  
Vol 110 (1) ◽  
pp. 186-192 ◽  
Author(s):  
Monica V. Goldberg ◽  
Charles H. Maris ◽  
Edward L. Hipkiss ◽  
Andrew S. Flies ◽  
Lijie Zhen ◽  
...  

Expression of the PD-1 receptor on T cells has been shown to provide an important inhibitory signal that down-modulates peripheral effector responses in normal tissues and tumors. Furthermore, PD-1 up-regulation on chronically activated T cells can maintain them in a partially reversible inactive state. The function of PD-1 in the very early stages of T-cell response to antigen in vivo has not been fully explored. In this study, we evaluate the role of PD-1 and its 2 B7 family ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), in early fate decisions of CD8 T cells. We show that CD8 T cells specific for influenza hemagglutinin (HA) expressed as a self-antigen become functionally tolerized and express high levels of surface PD-1 by the time of their first cell division. Blockade of PD-1 or B7-H1, but not B7-DC, at the time of self-antigen encounter mitigates tolerance induction and results in CD8 T-cell differentiation into functional cytolytic T lymphocytes (CTLs). These findings demonstrate that, in addition to modulating effector functions in the periphery, B7-H1:PD-1 interactions regulate early T-cell–fate decisions.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4533-4533
Author(s):  
Ying Wang ◽  
Shan He ◽  
Yongnian Liu ◽  
Robert Hooper ◽  
Hongshuang Yu ◽  
...  

Abstract Graft-versus-host disease (GVHD) remains a major barrier for the success of allogeneic hematopoietic stem cell transplantation (allo-HSCT). We have identified the central role of the histone methyltransferase Ezh2 in regulating allogeneic T-cell expansion, differentiation and function. Conditional loss of Ezh2 in donor T cells inhibits GVHD in mice due to the inability of alloreactive T cells to persist. However, the molecular mechanism by which Ezh2 deficiency causes alloreactive T cell death remains unknown. Here we demonstrate that genetic deletion of Stromal Interaction Molecule (Stim) 1, a dynamic endoplasmic reticulum Ca2+ sensor and regulator of Ca2+ signaling, rescues antigen-activated Ezh2-null (Ezh2-/-) T cells, leading to restored persistence of alloreactive effector T cells in mice and severe GVHD. Using RNA-sequencing analysis, we found Ezh2-deficiency led to the upregulation of multiple genes (e.g., Ifng, Prf1, Ccl5, Ccl4, Upp1 and Spp1) known to be regulated by Ca2+ signals through calcineurin (CN), the primary target of the immunosuppressant cyclosporine A (CsA). This reverse correlation between Ezh2 inhibition and CsA-treatment for gene expression suggests that Ezh2 may antagonize Ca2+ signaling in activated T cells. Calcium signaling assays revealed higher cytosolic Ca2+ uptake and more frequent Ca2+ oscillations in Ezh2-/- T cells. Moreover, Ezh2-/- T cells exhibited significantly increased polarization of Stim1 and Orai1 in the cellular membrane. These data reveal an unexpected role of Ezh2 as a negative regulator of Ca2+ entry, thereby serving as a 'brake' for Ca2+ signaling. Using the C57BL/6 (B6) into Balb/c mouse GVHD model, we found significantly fewer Ezh2-/- or Stim1-/- IFN-g-secreting effector T cells compared to the WT counterparts on day 8 or 14 post-transplantation. In contrast, deleting Stim1 from Ezh2-/- donor T cells rescued the cells in the spleen and liver, producing even more donor T cells and IFN-g-secreting effector T cells compared to WT T cells and inducing severe GVHD. We further examined the cell autonomous effect of Stim1 deletion on the rescue of Ezh2-/- T cells by mixing WT T cells (B6/SJL, CD45.1) with Ezh2- and/or Stim1- conditional knockout T cells (i.e., Ezh2-/-, Stim1-/- or Ezh2-/- x Stim1-/- B6 T cells (CD45.2)) at a ratio of 1:1 before transferring into the Balb/c mice. While loss of either Ezh2 or Stim1 led to lower frequency of IFN-g+IL-2+ effector T cells, combined deletion of both genes restored the frequency and number of IFN-g+IL-2+ effector T cells to that of WT T cells. Thus, Stim1-mediated Ca2+ signals are crucial for mediating cell death in alloantigen-driven Ezh2-/- effector T cells. To further determine whether the inhibition of CN-NFAT contributes to the rescue, we treated T cell receptor (TCR)-activated Ezh2-/- T cells with CsA or the calcium release-activated channel specific inhibitor BTP2, respectively, in vitro. While BTP2 dramatically improved the survival of IFN-g-producing effector T cells, CsA did not, suggesting the involvement of CN-NFAT-independent pathways. Ca2+ overload is known to impair mitochondrial function and cause massive cell death. As compared to TCR-activated WT T cells, activated Ezh2-/- T cells displayed significantly less ATP, lower mitochondrial membrane potential, enlarged mitochondrial mass, and decreased capacity to upregulate oxidative phosphorylation. Stim1 deletion largely reversed the metabolic defect in Ezh2-/- T cells, indicating the critical role of mitochondrial metabolism in rescuing these T cells. Considered together, our findings identify the remarkable coordination between Ezh2- and Stim1-regulated effector T cell persistence. As such, these investigations may lead to new approaches to inhibit GVHD, with broad implications to defining fundamental mechanisms of T cell differentiation for control of adaptive immunity, such as tumor immunity and autoimmunity. Disclosures Reshef: Incyte: Consultancy; Takeda Pharmaceuticals: Consultancy; Pfizer: Consultancy; Kite Pharma: Consultancy; Atara Biotherapeutics: Consultancy; Bristol-Myers Squibb: Consultancy.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4228-4228
Author(s):  
Wendy Kandell ◽  
Thu Le Trinh ◽  
Xianghong Chen ◽  
Pingyan Cheng ◽  
Danielle Gilvary ◽  
...  

We have previously reported that the overexpression of S100A9 drives the development of Myelodysplastic Syndrome (MDS) through expansion of Myeloid Derived Suppressor Cells (MDSC) and promotion of pyroptosis. Despite the identified role of S100A9's effects on MDSC, and hematopoietic stem and progenitor cells (HSPC), as well as their establishment of an immunosuppressive microenvironment, the effects of S100A9 on adaptive immunity in MDS progression are less clear. Here, we report for the first time the unidentified role of S100A9 on T cell function in MDS that may lead to impaired immunosurveillance in the disease. Danger Associated Molecular Pattern (DAMP) S100A9 is a known ligand for the Pattern Recognition Receptor (PRR) Receptor for Advanced Glycation Endproducts (RAGE). We investigated RAGE surface expression by flow cytometry on MDS bone marrow resident T cells vs those derived from healthy donor bone marrow. We found significantly (p=0.04) higher RAGE surface expression on T cells from MDS bone marrow, and this expression was restricted to the CD4 lineage. To ascertain the effects of S100A9 on RAGE+CD4+ T cell function, we performed flow cytometry in a time course experiment post-T cell activation. Without S100A9 treatment, cell surface RAGE expression was low in activated T cells from healthy donors, but extended treatment with recombinant human S100A9 resulted in increased RAGE expression, suggesting a positive feedback loop for this DAMP. Unlike activated T cells, T cells not exposed to activating conditions did not display upregulated RAGE expression after S100A9 treatment. This indicates that this may be a post-activation switch, acting as a checkpoint for the T cell in the context of excessive damage signaling by DAMP S100A9. In order to further characterize the functional consequences of RAGE engagement, we performed transcription factor staining paired with a cytometric bead array for secreted cytokines in activated T cells treated with S100A9. Tumor Necrosis Factor Alpha (TNFa), IL-10 and IL-6 were induced by S100A9, indicating perhaps some degree of polarization induced by this DAMP. Commercially available RAGE V-domain inhibitor FPS-ZM1 blunted this cytokine signaling, indicating a significant portion of this cytokine production is indeed mediated through RAGE. In addition, we performed lipophilic dye dilution assays to track the effects S100A9 has on T cell proliferation following activation. S100A9 significantly decreased proliferative response under normal stimulatory conditions. Similar inhibition was seen in T cells derived from PBMC, MDS, or healthy donor bone marrow resident T cells, suggesting that the consequences of RAGE engagement are not disease specific. To rule out apoptosis as a potential cause for this halt in proliferation, we stained the cells with Annexin V and Propidium Iodide. To further elucidate how S100A9 might be affecting T cell proliferation, we analyzed cell cycle profiles following activation and S100A9 treatment. T cells treated with S100A9 showed a repressed cell cycle prior to G2, compared to T cells activated without any S100A9 treatment, suggesting a possible G1/S arrest. The evidence obtained in our study suggests any role of T cell dysfunction mediated by RAGE in MDS may be directly linked to the increased levels of S100A9 in the bone marrow microenvironment. Our work represents a novel mechanism of T cell dysfunction that may lead to a lack of responsiveness in the context of a disease known to overexpress the RAGE ligand S100A9. Capitalizing on this novel checkpoint can potentially be used both as novel biomarker and as a therapeutic target in the future to restore T cell immunosurveillance to a functional state in MDS. Disclosures List: Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 342-342
Author(s):  
Yi Gu ◽  
Hee-Don Chae ◽  
Jamie E. Siefring ◽  
Aparna C. Jasti ◽  
David A. Hildeman ◽  
...  

Abstract Engagement of the T cell receptor (TCR) by antigen/major histocompatibility complex initiates a signaling cascade involving activation and recruitment of multiple key intracellular proteins, including the zeta-chain-associated protein 70 (ZAP-70). ZAP-70-deficient mice show thymocyte developmental arrest and mutations of ZAP-70 are associated with severe combined immunodeficiency in humans. However, regulation of these early biochemical processes is not completely understood. RhoH, first identified as a hypermutable gene in non-Hodgkin’s lymphomas, belongs to the family of Rho guanosine triphosphatases (Rho GTPases), which are recognized as critical mediators of signaling pathways regulating cell growth, differentiation and function (Gu et al., Science 302: 445-9). The expression of RhoH is restricted to hematopoietic cells, and using in vitro protein-binding and immunoprecipitation assays, we found that RhoH interacts with ZAP-70 in the TCR complex. Therefore, to understand the role of this highly expressed protein in T cell development and function, we have generated RhoH-deficient mice using homologous recombination. RhoH-/- mice are viable and normal in body size. However, the thymuses of 4–6 week-old RhoH-/- mice are significantly smaller and show a 3-fold decrease in cellularity compared with those from WT littermates. RhoH-/- thymocyte development is severely blocked at the CD4+ CD8+ double positive (DP) stage, resulting in markedly fewer CD4 or CD8 single positive (SP) T cells in RhoH-/- thymus (CD4 SP: 10.5+/− 1.6 vs. 1.1+/−0.2; CD8 SP: 6.8+/−0.7 vs. 0.8+/−0.1, cell number x106, WT vs. RhoH−/−, n≥15 mice, p<0.001). Numbers of peripheral T cells are also significantly reduced in RhoH−/− mice. RhoH DP thymocytes exhibit reduced surface expression of TCRb, but normal levels of TCRb mRNA. Further, the few circulating T cells in RhoH−/− mice exhibit normal expression of TCRb. These data suggest that reduced TCRb surface expression is a result, rather than a cause of the developmental arrest. In addition, expression of the activation marker, CD5, is severely reduced on RhoH−/− DP thymocytes (mean fluorescence intensity: 65.3+/−4.7 vs. 7.2+/−0.9, WT vs. RhoH−/−, n≥9 mice, p<0.01). These phenotypes are similar to ZAP-70-deficient mice and suggest that the thymocyte developmental arrest is due to defective TCR signaling pathway in RhoH−/− mice. To determine the potential role of RhoH in TCR signaling, we studied proliferative responses to anti-CD3 antibody cross-linking of RhoH−/− spleen T cells. RhoH−/− cells show significantly defective proliferation (3H-thymidine incorporation: 10.5+/−0.9 vs. 2.1+/−0.2, cpm x103, WT vs. RhoH−/−, n=6, p<0.01) as well as impaired survival (% viable cells: 27+/−2 vs. 6+/−1, WT vs. RhoH−/−, n=3, p<0.01). Additionally, activation of Rac GTPases and p42/p44 mitogen-activated protein kinases, known downstream effectors of the TCR-ZAP70 complex, is significantly reduced in response to TCR stimulation in RhoH−/− T cells as compared with WT cells. Finally, we show that retrovirus-mediated re-expression of RhoH into RhoH−/− bone marrow is sufficient to restore normal thymic development in Rag2−/− recipient mice, demonstrating the specific role of RhoH in thymic development. Together, our studies demonstrate that RhoH is essential for proper thymocyte development and function, and also implicate RhoH as a novel, critical regulator of T cell signaling possibly through its interaction with ZAP-70 in the TCR complex.


1983 ◽  
Vol 158 (4) ◽  
pp. 1077-1091 ◽  
Author(s):  
P Marrack ◽  
R Endres ◽  
R Shimonkevitz ◽  
A Zlotnik ◽  
D Dialynas ◽  
...  

We have examined the role of the murine homologue of Leu-3 T4, L3T4, in recognition of antigen in association with products of the major histocompatibility complex (Ag/MHC) by murine T cell hybridomas. A series of ovalbumin (OVA)/I-Ad-specific T cell hybridomas were ranked in their sensitivity to Ag/I by measuring their ability to respond to low doses of OVA, or their sensitivity to inhibition by anti-I-Ad antibodies. T cell hybridomas with low apparent avidity for OVA/I-Ad, i.e. that did not respond well to low concentrations of OVA and were easily inhibited by anti-I-Ad, were also easily inhibited by anti-L3T4 antibodies. The reverse was true for T cell hybridomas with apparent high avidity for Ag/MHC. We found that the presence of low doses of anti-L3T4 antibodies caused T cell hybridomas to respond less well to low doses of Ag, and to be more easily inhibited by anti-I-Ad antibodies. These results suggested that the role of the L3T4 molecule is to increase the overall avidity of the reaction between T cells and Ag-presenting cells. In support of this idea was the discovery of several L3T4- subclones of one of our L3T4+ T cell hybridomas, D0.11.10. The L3T4- subclones had the same amount of receptor for OVA/I-Ad as their L3T4+ parent, as detected by an anti-receptor monoclonal antibody. The L3T4- subclones, however, responded less well to low doses of OVA, and were more easily inhibited by anti-I-Ad antibodies than their L3T4/ parent. These results showed that the L3T4 molecule was not required for surface expression of, or functional activity of, the T cell receptor for Ag/MHC. The L3T4 molecule did, however, increase the sensitivity with which the T cell reacted with Ag/MHC on Ag-presenting cells.


1995 ◽  
Vol 181 (1) ◽  
pp. 71-77 ◽  
Author(s):  
M R Alderson ◽  
T W Tough ◽  
T Davis-Smith ◽  
S Braddy ◽  
B Falk ◽  
...  

A significant proportion of previously activated human T cells undergo apoptosis when triggered through the CD3/T cell receptor complex, a process termed activation-induced cell death (AICD). Ligation of Fas on activated T cells by either Fas antibodies or recombinant human Fas-ligand (Fas-L) also results in cytolysis. We demonstrate that these two pathways of apoptosis are causally related. Stimulation of previously activated T cells resulted in the expression of Fas-L mRNA and lysis of Fas-positive target cells. Fas-L antagonists inhibited AICD of T cell clones and staphylococcus enterotoxin B (SEB)-specific T cell lines. The data indicate AICD in previously stimulated T cells is mediated by Fas/Fas-L interactions.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A626-A626
Author(s):  
Annah Rolig ◽  
Daniel Rose ◽  
Grace Helen McGee ◽  
Saul Kivimae ◽  
Werner Rubas ◽  
...  

BackgroundTumor cell death caused by radiation therapy (RT) can trigger anti-tumor immune responses in part because dying cells release adjuvant factors that amplify and sustain DC and T cell responses. We previously demonstrated that bempegaldesleukin (BEMPEG:NKTR-214, a first-in-class CD122-preferential IL-2 pathway agonist), significantly enhanced the anti-tumor efficacy of RT through a T cell-dependent mechanism. Because RT can induce either immunogenic or tolerogenic cell death, depending on a multitude of factors (radiation dose, cell cycle phase, and tumor microenvironment), we hypothesized that providing a specific immunogenic adjuvant, like intratumoral NKTR-262, a novel toll-like receptor (TLR) 7/8 agonist, to the tumor site would further improve systemic tumor-specific immunity by promoting synergistic activation of local immunostimulatory innate immune responses. Therefore, we evaluated whether intratumoral NKTR-262, combined with systemic BEMPEG treatment would result in improved tumor-specific immunity and survival compared to BEMPEG combined with RT.MethodsTumor-bearing mice (CT26; EMT6) received BEMPEG (0.8 mg/kg; iv), RT (16 Gy x 1), and/or intratumoral NKTR-262 (0.5 mg/kg). Flow cytometry was used to evaluate CD4+ and CD8+ T cell activation status in the blood and tumor (7 days post-treatment). The contribution of specific immune subsets was determined by depletion of CD4+, CD8+, or NK cells. CD8+ T cell cytolytic activity was determined in vitro with an Incucyte assay. Data are representative of 1–2 independent experiments (n=5–14/group) and statistical significance was determined by 1-way ANOVA (p-value cut-off of 0.05).ResultsBEMPEG/NKTR-262 resulted in significantly improved survival compared to BEMPEG/RT. Both combination therapies were CD8+ T cell dependent. However, response to BEMPEG/NKTR-262 was characterized by a significant expansion of activated CD8+ T cells (GzmA+; Ki-67+; ICOS+; PD-1+) in the blood, which correlated with reduced tumor size (p<0.05). In the tumor, BEMPEG/NKTR-262 induced higher frequencies of GzmA+ CD8+ T cells exhibiting reduced expression of suppressive molecules (PD-1+, TIM-3+), compared to BEMPEG/RT. Additionally, CD8+ T cells isolated from BEMPEG/NKTR-262-treated tumors had greater cytolytic capacity than those from BEMPEG/RT-treated mice.ConclusionsCombining BEMPEG with NKTR-262 lead to a more robust expansion of activated CD8+ T cells compared to the BEMPEG/RT combination. Enhancement of the activated CD8+ T cell response in mice treated with NKTR-262 in combination with BEMPEG suggests that intratumoral TLR stimulation provides superior antigen presentation and costimulatory activity compared to RT. A clinical trial of BEMPEG/NKTR-262 for patients with metastatic solid tumors is in progress (NCT03435640).


2016 ◽  
Vol 36 (24) ◽  
pp. 3113-3127 ◽  
Author(s):  
Martin G. Sauer ◽  
Jessica Herbst ◽  
Ulf Diekmann ◽  
Christopher E. Rudd ◽  
Christian Kardinal

The clinical potential of transplantation is often reduced by T cell-mediated alloresponses that cause graft rejection or graft-versus-host disease. Integrin-mediated adhesion between alloreactive T cells and antigen-presenting cells is essential for allorejection. The identity of the signaling events needed for the activation of integrins such as LFA-1 is poorly understood. Here, we identified a novel role of the protein tyrosine phosphatase SHP-1 in the regulation of murine LFA-1-mediated adhesion in an allograft setting. Upon alloactivation, SHP-1 activity is reduced, resulting in an increase in LFA-1 adhesion compared to that for syngeneically activated T cells. The importance of these differential activation properties was further indicated by small interfering RNA (siRNA) knockdown of SHP-1 in syngeneically and allogeneically stimulated T cells. Mechanistically, SHP-1 modulated the binding of SLP-76 to ADAP by dephosphorylation of the YDGI tyrosine motif of ADAP, a known docking site for the Src family kinase Fyn. This novel key role of SHP-1 in the regulation of LFA-1-mediated adhesion may provide a new insight into T cell-mediated alloresponses and may pave the way to the development of new immunosuppressive pharmaceutical agents.


Sign in / Sign up

Export Citation Format

Share Document