Regulation of Tim-3 function by binding to phosphatidylserine

2021 ◽  
Vol 478 (22) ◽  
pp. 3999-4004
Author(s):  
Lawrence P. Kane

Tim-3 is a transmembrane protein that is highly expressed on subsets of chronically stimulated CD4+ helper and CD8+ cytotoxic T cells, with more transient expression during acute activation and infection. Tim-3 is also constitutively expressed by multiple types of myeloid cells. Like other TIM family members, Tim-3 can bind to phosphatidylserine displayed by apoptotic cells, and this interaction has been shown to mediate uptake of such cells by dendritic cells and cross-presentation of antigens to CD8+ T cells. In contrast, how the recognition of PS by Tim-3 might regulate the function of Tim-3+ T cells is not known. In their recent paper, Lemmon and colleagues demonstrate for the first time that recognition of PS by Tim-3 leads to enhanced T cell activation.

2020 ◽  
Vol 18 (1) ◽  
Author(s):  
Jean-Philippe Bastien ◽  
Natalie Fekete ◽  
Ariane V. Beland ◽  
Marie-Paule Lachambre ◽  
Veronique Laforte ◽  
...  

Abstract Background A major obstacle to anti-viral and -tumor cell vaccination and T cell immunotherapy is the ability to produce dendritic cells (DCs) in a suitable clinical setting. It is imperative to develop closed cell culture systems to accelerate the translation of promising DC-based cell therapy products to the clinic. The objective of this study was to investigate whether viral antigen-loaded monocyte-derived DCs (Mo-DCs) capable of eliciting specific T cell activation can be manufactured in fluorinated ethylene propylene (FEP) bags. Methods Mo-DCs were generated through a protocol applying cytokine cocktails combined with lipopolysaccharide or with a CMV viral peptide antigen in conventional tissue culture polystyrene (TCPS) or FEP culture vessels. Research-scale (< 10 mL) FEP bags were implemented to increase R&D throughput. DC surface marker profiles, cytokine production, and ability to activate antigen-specific cytotoxic T cells were characterized. Results Monocyte differentiation into Mo-DCs led to the loss of CD14 expression with concomitant upregulation of CD80, CD83 and CD86. Significantly increased levels of IL-10 and IL-12 were observed after maturation on day 9. Antigen-pulsed Mo-DCs activated antigen-responsive CD8+ cytotoxic T cells. No significant differences in surface marker expression or tetramer-specific T cell activating potency of Mo-DCs were observed between TCPS and FEP culture vessels. Conclusions Our findings demonstrate that viral antigen-loaded Mo-DCs produced in downscaled FEP bags can elicit specific T cell responses. In view of the dire clinical need for closed system DC manufacturing, FEP bags represent an attractive option to accelerate the translation of promising emerging DC-based immunotherapies.


2018 ◽  
Vol 36 (5_suppl) ◽  
pp. 6-6
Author(s):  
Stephanie Dorta-Estremera ◽  
Krishna Nookala Sita Mahalakshmi ◽  
Ananta V Yanamandra ◽  
Lauren Elizabeth Colbert ◽  
Guojun Yang ◽  
...  

6 Background: Limited data in cancer patients have suggested that chemotherapy and radiation impact local and systemic immune cell populations. Radiation therapy (RT) is known to deplete circulating lymphocytes but is thought to increase local antigen presentation. The dynamics of these competing effects on the kinetics of intratumoral infiltration and expansion of activated and immunoregulatory T cells are unknown. Methods: We prospectively evaluated intratumoral immune infiltration during fractionated RT using multi-spectral flow cytometry. Cervical brushings were obtained from 14 patients before (baseline) and during RT (week 1, 3 and 5). Cells collected from the cervical brushings were stained with a 16-color panel of antibodies that included markers to identify T cell and dendritic cell subsets with activation and suppressor molecules. Changes in immune cell subsets at different time points were evaluated and calculated using matched-pair analysis with Wilcoxon rank sum test. Results: CD3+ T cells declined over the first week of treatment (28% of CD3 at baseline, vs. 14.8% at week 1, p = 0.0273). The percentage of CD3+ cells subsequently increased at 3 weeks (25.6%) and 5 weeks (37.8%). Both CD8+ and CD4+ T cells underwent a decline at week 1 followed by expansion at week 3 and 5. Percentages of regulatory T cells (CD4+Foxp3+) showed a similar trend of reduction and further expansion but did not reach significance. The percentage of CD8+ T cells expressing the T cell activation marker CD69 and the cytotoxic protease Granzyme B (GrzB) continuously increased over time (CD69+: 11.8%, 27.7%, 38.7%, 57.5%, and GrzB+: 23.9%, 53.2%, 48.1%, 58.2%). While the percentage of dendritic cells (CD11c+ CD11b+) was stable during treatment, the subset of activated dendritic cells expressing CD86 increased at week 1 and subsequently declined (week 1, 19.1% vs week 5, 9.8%, p = 0.0642). Conclusions: Activated CD8+ effector T cells expand in the cervix during radiation therapy. Moreover, in the first week of treatment, CD8+ T cells contract while dendritic cells undergo activation suggesting this may be a critical time to intervene to maximize anti-tumor immunity.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A38.1-A38
Author(s):  
S Schmitt ◽  
A Lohner ◽  
K Deiser ◽  
A Maiser ◽  
M Rothe ◽  
...  

BackgroundDendritic cells (DCs) are antigen-presenting cells that induce antigen-specific T-cell responses. Therefore, they are used as tools and targets for anti-tumor vaccination. In contrast to T-cell based immunotherapies, that are often limited to surface antigens, DC-based vaccination strategies open up new therapeutic options by utilizing highly abundant intracellular tumor antigens as a target source. Among those, recent interest has been focused on the identification of neoantigens derived from tumor-specific mutations. Especially mutated Nucleophosmin 1 (ΔNPM1) is a considered candidate for targeted therapy in acute myeloid leukemia (AML). We developed a multifunctional antibody construct consisting of a peptide domain including a variable T-cell epitope that is fused to an αCD40 single chain variable fragment (scFv) with agonistic function to target and activate dendritic cells in vivo. To potentiate therapeutic efficacy, toll-like receptor (TLR) agonists can be attached as co-stimulatory domains, thereby aiming to enhance cross-presentation of conjugated (neo)antigens to CD8+ T cells.Materials and MethodsFlow cytometry and microscopy-based binding and internalization experiments were performed using monocyte-derived dendritic cells (moDCs). Upregulation of surface markers (CD80, CD83, CD86, HLA-DR) as well as cytokine secretion (IL-6 and IL-12) indicated DC maturation. To validate peptide processing and presentation, moDCs were co-cultured with autologous as well as allogeneic T cells. IFN-γ and TNF-α secretion served as a readout for T-cell activation, peptide-MHC multimer staining for T-cell proliferation.ResultsFor proof-of-principle experiments, the multispecific antibody derivative was developed by fusing the αCD40 scFv to a cytomegalovirus (CMV)-specific peptide. The αCD40.CMV construct bound CD40 agonistically and showed efficient internalization into early endosomal compartments on immature moDCs. In co-cultures of immature and mature moDCs with autologous or allogeneic T cells, αCD40.CMV induced a significantly increased T-cell activation and proliferation compared to the control. The co-administration of αCD40.CMV with various TLR agonists as vaccine adjuvants resulted in a significant upregulation of DC maturation markers in comparison to αCD40.CMV only. Interestingly, not all adjuvants were able to enhance the T-cell response. To translate this principle to the AML setting, the CMV peptide sequence was replaced with the ΔNPM1-derived and HLA-A*02:01-binding neoantigen CLAVEEVSL. Cross-presentation to CD8+ T cells transduced with a ΔNPM1-specific T-cell receptor was proven by IFN-γ and TNF-α secretion in co-cultures with moDCs that have been pre-incubated with αCD40.ΔNPM1. The optimal vaccine adjuvant has yet to be identified.ConclusionsWe successfully demonstrated the development of a multifunctional antibody construct that specifically targets and stimulates DCs by an agonistic αCD40 scFv. It simultaneously delivers a T cell-specific peptide with a vaccine adjuvant to induce an efficient T-cell response. As neoantigens are promising targets and under intense investigaton, the αCD40.ΔNPM1 fusion protein is of high therapeutic interest. Thus, our approach displays a promising DC vaccination option for the treatment of AML.Disclosure InformationS. Schmitt: None. A. Lohner: None. K. Deiser: None. A. Maiser: None. M. Rothe: None. C. Augsberger: None. A. Moosmann: None. H. Leonhardt: None. N. Fenn: None. M. Griffioen: None. K. Hopfner: None. M. Subklewe: None.


Blood ◽  
2013 ◽  
Vol 121 (26) ◽  
pp. 5184-5191 ◽  
Author(s):  
Catherine E. Terrell ◽  
Michael B. Jordan

Key PointsDefects in perforin and related genes lead to abnormal T-cell activation and are associated with HLH. The physiological mechanism by which perforin protects from HLH involves CD8+ T-cell elimination of rare antigen-presenting dendritic cells.


2007 ◽  
Vol 83 (3) ◽  
pp. 742-754 ◽  
Author(s):  
Masashi Takahara ◽  
Manami Miyai ◽  
Mai Tomiyama ◽  
Masato Mutou ◽  
Andrew J. Nicol ◽  
...  

2021 ◽  
Vol 17 (7) ◽  
pp. e1009771
Author(s):  
Ondrej Cerny ◽  
Camilla Godlee ◽  
Romina Tocci ◽  
Nancy E. Cross ◽  
Haoran Shi ◽  
...  

The Salmonella enterica effector SteD depletes mature MHC class II (mMHCII) molecules from the surface of infected antigen-presenting cells through ubiquitination of the cytoplasmic tail of the mMHCII β chain. This requires the Nedd4 family HECT E3 ubiquitin ligase Wwp2 and a tumor-suppressing transmembrane protein adaptor Tmem127. Here, through a proteomic screen of dendritic cells, we found that SteD targets the plasma membrane protein CD97 for degradation by a similar mechanism. SteD enhanced ubiquitination of CD97 on K555 and mutation of this residue eliminated the effect of SteD on CD97 surface levels. We showed that CD97 localises to and stabilises the immunological synapse between dendritic cells and T cells. Removal of CD97 by SteD inhibited dendritic cell-T cell interactions and reduced T cell activation, independently of its effect on MHCII. Therefore, SteD suppresses T cell immunity by two distinct processes.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 992-992 ◽  
Author(s):  
Steven Coles ◽  
Marie N Gilmore ◽  
Reiss Reid ◽  
Steven Knapper ◽  
Alan K. Burnett ◽  
...  

Abstract Long-term remission for acute myeloid leukemia (AML) is still not achieved for the majority of patients and consequently there is a need for new treatments to consolidate current therapy. A promising approach is to augment the anti-tumor immune response in these patients; however most cancers do not activate immune effector cells because they express immunosuppressive ligands. Previously we showed that CD200 overexpression on AML blasts suppresses memory CD4+ and CD8+ T cell effector function through engagement with CD200 receptor (CD200R) on these cells. Blocking CD200:CD200R, however, only partially restored T cell activity, suggesting that alternative immunosuppressive mechanisms were involved. Recently, promising clinical outcomes have been reported for melanoma and non-small cell lung cancer using humanized antibodies targeting another immunosuppressive receptor, PD-1, and we therefore investigated whether this could be contributing to the immunosuppression of T cell effector responses in CD200hi AML patients. Initially, we investigated whether CD200 and the immunosuppressive ligand for PD-1, PD1-L1, were co-expressed in AML blasts at diagnosis. Affymetrix gene expression data from 158 AML blasts showed that AML patients in the upper quartile for CD200 expression (CD200hi) had 10-fold higher levels of PD1-L1 expression compared to CD200lo (lower quartile) patients. Analysis of CD200 and PD1-L1 protein expression on AML blast cells confirmed this association at the protein level (r2 = 0.49; p<0.01). The co-expression of CD200 and PD1-L1 on patient AML blast cells, suggested that they cooperated in immunosuppression. In support of this, we found that the CD200 and PD1-L1 cognate co-receptors (CD200R and PD-1 respectively) were present on CD4+ and CD8+ T cells from AML patients. Further characterization of PD-1+ T cells showed that the mean frequency of PD-1+ early differentiated T cells (CD57- CD28+) was increased for CD200hi AML patients CD4+ (19% ± 3 vs 13% ± 3; p<0.05) and CD8+ T cells (21% ± 3 vs 11% ± 2; p<0.05). We also found that the mean frequency of late differentiated CD8+ T cells that have poor anti-tumor function (CD57+ CD28- PD-1+) was almost twice that for CD200hi patients compared with CD200lo (38% ± 6 vs 21% ± 9 respectively; p<0.05). Expansion of these cells was also associated with a decreased CD4:CD8 ratio in these patients (2.1 ± 0.5 vs 3.7 ± 1 for CD200hi and CD200lo respectively; p<0.01). . These findings show for the first time a link between CD200 expression level on AML blast cells and the frequency of PD-1+ late differentiated CD8+ T cells. To directly test whether engagement of CD200 with CD200R was capable of mediating PD-1 up-regulation on CD8+ T cells, we co-cultured a CD8+ CD200R+ T cell clone (7E7) either with K562 cells stably overexpressing CD200 or K562 empty vector controls (negative for CD200). Co-culture with CD200+ cells, significantly increased the frequency of PD-1+ T cells (26% ± 3 vs 17% ± 4; p<0.05) and this was antagonized by CD200 blocking antibody (26% ± 3 vs 21% ± 3; p<0.01). These data show that CD200:CD200R interaction has the capacity to increase the frequency of PD-1+ CD8+ T cells. To model the functional implications of this, we created a series of K562 lines expressing CD200, PD1-L1 or both molecules in combination and analyzed the effect of T cell activation (via TNFα production). We found that both CD200 and PD1-L1 induced a similar (>50%) reduction in the frequency of activated 7E7 T cells; however, when both CD200 and PD1-L1 were co-expressed, T cell activation was almost ablated (~90% reduction; p<0.01). Moreover, the strength of the TNFα response was also reduced in co-culture assays where either CD200 or PD1-L1 were present, indicating a direct effect at the level of CD8+ T cell function (2.8 ± 0.5 vs 1.7 ± 0.5; p<0.05). These data demonstrate that CD200:CD200R and PD1-L1:PD-1 engagement on T cells can act in tandem to augment immunosuppression of CD8+ T cells. In summary, we show for the first time that the immunosuppressive molecules, CD200 and PD1-L1 appear to be co-regulated on AML blasts and that these can act in combination to profoundly suppress T cell activation. Further, we show that CD200:CD200R engagement induces PD-1+ CD8+ T cells. Taken together we propose a novel CD200/PD1-L1 immunotherapeutic synapse in AML which should be targeted by combining CD200:CD200R and PD1-L1:PD-1 blockade in immunotherapy of AML. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 6 (57) ◽  
pp. eabf7570
Author(s):  
Laura A. Vella ◽  
Josephine R. Giles ◽  
Amy E. Baxter ◽  
Derek A. Oldridge ◽  
Caroline Diorio ◽  
...  

Pediatric COVID-19 following SARS-CoV-2 infection is associated with fewer hospitalizations and often milder disease than in adults. A subset of children, however, present with Multisystem Inflammatory Syndrome in Children (MIS-C) that can lead to vascular complications and shock, but rarely death. The immune features of MIS-C compared to pediatric COVID-19 or adult disease remain poorly understood. We analyzed peripheral blood immune responses in hospitalized SARS-CoV-2 infected pediatric patients (pediatric COVID-19) and patients with MIS-C. MIS-C patients had patterns of T cell-biased lymphopenia and T cell activation similar to severely ill adults, and all patients with MIS-C had SARS-CoV-2 spike-specific antibodies at admission. A distinct feature of MIS-C patients was robust activation of vascular patrolling CX3CR1+ CD8+ T cells that correlated with the use of vasoactive medication. Finally, whereas pediatric COVID-19 patients with acute respiratory distress syndrome (ARDS) had sustained immune activation, MIS-C patients displayed clinical improvement over time, concomitant with decreasing immune activation. Thus, non-MIS-C versus MIS-C SARS-CoV-2 associated illnesses are characterized by divergent immune signatures that are temporally distinct from one another and implicate CD8+ T cells in the clinical presentation and trajectory of MIS-C.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii96-ii96
Author(s):  
Catalina Lee Chang ◽  
Jason Miska ◽  
David Hou ◽  
Aida Rashidi ◽  
Peng Zhang ◽  
...  

Abstract Immunotherapy has revolutionized the treatment of many tumors. However, most glioblastoma (GBM) patients have not, so far, benefited from such successes. With the goal of exploring ways to boost anti-GBM immunity, we developed a B-cell-based vaccine (BVax) that consists of 4-1BBL+ B cells activated with CD40 agonism and IFNg stimulation. BVaxmigrate to key secondary lymphoid organs and are proficient at antigen cross-presentation, which promotes both the survival and functionality of CD8+ T cells. A combination of radiation, BVax, and PD-L1 blockade conferred tumor eradication in 80% of treated tumor-bearing animals. This treatment elicited immunologic memory that prevented the growth of new tumors upon subsequent re-injection in cured mice. GBM patient-derived BVax were successful in activating autologous CD8+ T cells; these T cells showed a strong ability to kill autologous glioma cells. In addition to the role in activating CD8+ T cells, BVax produce tumor-specific antibodies able to control tumor growth via antibody-mediated cell cytotoxicity. In conclusion, BVax tackles GBM immunosurveillance escape by using both cellular (CD8+ T-cell activation) and humoral (anti-tumor antibody production) immunity. Our study provides an efficient alternative to current immunotherapeutic approaches that can be readily translated to the clinic.


2000 ◽  
Vol 165 (8) ◽  
pp. 4305-4311 ◽  
Author(s):  
Gérard Eberl ◽  
Pierre Brawand ◽  
H. Robson MacDonald

Sign in / Sign up

Export Citation Format

Share Document