Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Nat Immunol ; 25(5): 802-819, 2024 May.
Article in English | MEDLINE | ID: mdl-38684922

ABSTRACT

Sepsis induces immune alterations, which last for months after the resolution of illness. The effect of this immunological reprogramming on the risk of developing cancer is unclear. Here we use a national claims database to show that sepsis survivors had a lower cumulative incidence of cancers than matched nonsevere infection survivors. We identify a chemokine network released from sepsis-trained resident macrophages that triggers tissue residency of T cells via CCR2 and CXCR6 stimulations as the immune mechanism responsible for this decreased risk of de novo tumor development after sepsis cure. While nonseptic inflammation does not provoke this network, laminarin injection could therapeutically reproduce the protective sepsis effect. This chemokine network and CXCR6 tissue-resident T cell accumulation were detected in humans with sepsis and were associated with prolonged survival in humans with cancer. These findings identify a therapeutically relevant antitumor consequence of sepsis-induced trained immunity.


Subject(s)
Macrophages , Neoplasms , Sepsis , Humans , Sepsis/immunology , Macrophages/immunology , Female , Neoplasms/immunology , Neoplasms/therapy , Male , Receptors, CXCR6/metabolism , Animals , T-Lymphocytes/immunology , Receptors, CCR2/metabolism , Middle Aged , Mice , Aged , Chemokines/metabolism , Adult
2.
Immunity ; 50(4): 1026-1028, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995494

ABSTRACT

Invariant Vγ9Vδ2 T cells respond to "phosphoantigen" metabolites through binding to the B30.2 domain of butyrophilin BTN3A. Yang et al. (2019) use molecular dynamic simulations based on X-ray structures of distinct B30.2 domain dimers to identify the asymmetric dimer as most active, which has implications for the inside-out signaling mechanism.


Subject(s)
Antigens, CD , Lymphocyte Activation , Butyrophilins , T-Lymphocytes/immunology , X-Rays
3.
Eur J Immunol ; 54(8): e2350773, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38804118

ABSTRACT

In the last decade, there has been a surge in developing immunotherapies to enhance the immune system's ability to eliminate tumor cells. Bispecific antibodies known as T cell engagers (TCEs) present an attractive strategy in this pursuit. TCEs aim to guide cytotoxic T cells toward tumor cells, thereby inducing a strong activation and subsequent tumor cell lysis. In this study, we investigated the activity of different TCEs on both conventional alpha-beta (αß) T cells and unconventional gamma delta (γδ) T cells. TCEs were built using camelid single-domain antibodies (VHHs) targeting the tumor-associated antigen CEACAM5 (CEA), together with T cell receptor chains or a CD3 domain. We show that Vγ9Vδ2 T cells display stronger in vitro antitumor activity than αß T cells when stimulated with a CD3xCEA TCE. Furthermore, restricting the activation of fresh human peripheral T cells to Vγ9Vδ2 T cells limited the production of protumor factors and proinflammatory cytokines, commonly associated with toxicity in patients. Taken together, our findings provide further insights that γδ T cell-specific TCEs hold promise as specific, effective, and potentially safe molecules to improve antitumor immunotherapies.


Subject(s)
Antibodies, Bispecific , Lymphocyte Activation , Receptors, Antigen, T-Cell, gamma-delta , Humans , Animals , Receptors, Antigen, T-Cell, gamma-delta/immunology , Antibodies, Bispecific/immunology , Lymphocyte Activation/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Single-Domain Antibodies/immunology , Cell Line, Tumor , T-Lymphocytes, Cytotoxic/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Carcinoembryonic Antigen/immunology
4.
Am J Hematol ; 99(3): 350-359, 2024 03.
Article in English | MEDLINE | ID: mdl-38165016

ABSTRACT

The presence of donor Vγ9Vδ2 T-cells after haploidentical hematopoietic stem cell transplant (h-HSCT) has been associated with improved disease-free survival. These cells kill tumor cells in a non-MHC restricted manner, do not induce graft-versus-host disease (GVHD), and can be generated by stimulation with zoledronic acid (ZA) in combination with interleukin-2 (IL-2). This monocentric phase I, open-label, dose-escalating study (ClinicalTrials.gov: NCT03862833) aimed at evaluating the safety and possibility to generate Vγ9Vδ2 T-cells early after h-HSCT. It applied a standard 3 + 3 protocol to determine the maximum tolerated dose (MTD) of increasing low-doses of IL-2 (5 days [d] per week, 4 weeks) in combination with a single dose of ZA, starting both the first Monday after d + 15 posttransplant. Vγ9Vδ2 T-cell monitoring was performed by multiparameter flow cytometry on blood samples and compared with a control cohort of h-HSCT recipients. Twenty-six patients were included between April 2019 and September 2022, 16 of whom being ultimately treated and seven being controls who received h-HSCT only. At the three dose levels tested, 1, 0, and 1 dose-limiting toxicities were observed. MTD was not reached. A significantly higher number of Vγ9Vδ2 T-cells was observed during IL-2 treatment compared with controls. In conclusion, early in vivo generation of Vγ9Vδ2 T-cells is feasible after h-HSCT by using a combination of ZA and repeated IL-2 infusions. This study paves the way to a future phase 2 study, with the hope to document lesser posttransplant relapse with this particular adaptive immunotherapy.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Hematopoietic Stem Cell Transplantation/methods , Interleukin-2 , Zoledronic Acid , T-Lymphocytes/pathology , Cyclophosphamide/therapeutic use , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Graft vs Host Disease/drug therapy , Stem Cell Transplantation
5.
Immunol Rev ; 298(1): 117-133, 2020 11.
Article in English | MEDLINE | ID: mdl-32965719

ABSTRACT

Despite recent significant progress in cancer immunotherapies based on adoptive cell transfer(s)(ACT), the eradication of cancers still represents a major clinical challenge. In particular, the efficacy of current ACT-based therapies against solid tumors is dramatically reduced by physical barriers that prevent tumor infiltration of adoptively transferred effectors, and the tumor environment that suppress their anti-tumor functions. Novel immunotherapeutic strategies are thus needed to circumvent these issues. Human peripheral blood Vγ9Vδ2 T cells, a non-alloreactive innate-like T lymphocyte subset, recently proved to be a promising anti-tumor effector subset for ACT-based immunotherapies. Furthermore, new cell engineering tools that leverage the potential of CRISPR/Cas technology open astounding opportunities to optimize their anti-tumor effector functions. In this review, we present the current ACT strategies based on engineered T cells and their limitations. We then discuss the potential of engineered Vγ9Vδ2 T cell to overcome these limitations and improve ACT-based cancer immunotherapies.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Humans , Neoplasms/therapy , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocyte Subsets
6.
Immunity ; 40(4): 490-500, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24703779

ABSTRACT

In humans, Vγ9Vδ2 T cells detect tumor cells and microbial infections, including Mycobacterium tuberculosis, through recognition of small pyrophosphate containing organic molecules known as phosphoantigens (pAgs). Key to pAg-mediated activation of Vγ9Vδ2 T cells is the butyrophilin 3A1 (BTN3A1) protein that contains an intracellular B30.2 domain critical to pAg reactivity. Here, we have demonstrated through structural, biophysical, and functional approaches that the intracellular B30.2 domain of BTN3A1 directly binds pAg through a positively charged surface pocket. Charge reversal of pocket residues abrogates binding and Vγ9Vδ2 T cell activation. We have also identified a gain-of-function mutation within this pocket that, when introduced into the B30.2 domain of the nonstimulatory BTN3A3 isoform, transfers pAg binding ability and Vγ9Vδ2 T cell activation. These studies demonstrate that internal sensing of changes in pAg metabolite concentrations by BTN3A1 molecules is a critical step in Vγ9Vδ2 T cell detection of infection and tumorigenesis.


Subject(s)
Antigens, CD/immunology , T-Lymphocytes/immunology , Antigens/immunology , Antigens, CD/chemistry , Antigens, CD/genetics , Butyrophilins , Cells, Cultured , Diphosphonates/immunology , Humans , Imidazoles/immunology , Intracellular Space , Lymphocyte Activation/genetics , Mutation/genetics , Protein Binding/genetics , Protein Engineering , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Structure, Tertiary/genetics , RNA, Small Interfering/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Zoledronic Acid
7.
Eur J Immunol ; 50(10): 1454-1467, 2020 10.
Article in English | MEDLINE | ID: mdl-32460359

ABSTRACT

Invariant Natural Killer T (iNKT) cells are particular T lymphocytes at the frontier between innate and adaptative immunities. They participate in the elimination of pathogens or tumor cells, but also in the development of allergic reactions and autoimmune diseases. From their first descriptions, the phenomenon of self-reactivity has been described. Indeed, they are able to recognize exogenous and endogenous lipids. However, the mechanisms underlying the self-reactivity are still largely unknown, particularly in humans. Using a CD1d tetramer-based sensitive immunomagnetic approach, we generated self-reactive iNKT cell lines from blood circulating iNKT cells of healthy donors. Analysis of their functional characteristics in vitro showed that these cells recognized endogenous lipids presented by CD1d molecules through their TCR that do not correspond to α-glycosylceramides. TCR sequencing and transcriptomic analysis of T cell clones revealed that a particular TCR signature and an expression of the SYK protein kinase were two mechanisms supporting human iNKT self-reactivity. The SYK expression, strong in the most self-reactive iNKT clones and variable in ex vivo isolated iNKT cells, seems to decrease the activation threshold of iNKT cells and increase their overall antigenic sensitivity. This study indicates that a modulation of the TCR intracellular signal contributes to iNKT self-reactivity.


Subject(s)
Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/genetics , Syk Kinase/metabolism , Animals , Antigens, CD1d/metabolism , Autoantigens/immunology , Autoimmunity , Cell Line , Humans , Lipids/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Syk Kinase/genetics , Transcriptome
8.
Proc Natl Acad Sci U S A ; 114(12): 3163-3168, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28270598

ABSTRACT

Human γδ T cells comprise a first line of defense through T-cell receptor (TCR) recognition of stressed cells. However, the molecular determinants and stress pathways involved in this recognition are largely unknown. Here we show that exposure of tumor cells to various stress situations led to tumor cell recognition by a Vγ8Vδ3 TCR. Using a strategy that we previously developed to identify antigenic ligands of γδ TCRs, annexin A2 was identified as the direct ligand of Vγ8Vδ3 TCR, and was found to be expressed on tumor cells upon the stress situations tested in a reactive oxygen species-dependent manner. Moreover, purified annexin A2 was able to stimulate the proliferation of a Vδ2neg γδ T-cell subset within peripheral blood mononuclear cells and other annexin A2-specific Vδ2neg γδ T-cell clones could be derived from peripheral blood mononuclear cells. We thus propose membrane exposure of annexin A2 as an oxidative stress signal for some Vδ2neg γδ T cells that could be involved in an adaptive stress surveillance.


Subject(s)
Annexin A2/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction , Stress, Physiological , T-Lymphocyte Subsets/metabolism , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/metabolism , Humans , Immunity, Innate , Ligands , Lymphocyte Activation , Neoplasms/immunology , Neoplasms/metabolism , Oxidative Stress , Protein Binding , Receptors, Antigen, T-Cell, gamma-delta/antagonists & inhibitors
9.
J Immunol ; 198(11): 4228-4234, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28461569

ABSTRACT

Vγ9Vδ2 T lymphocytes are the major human peripheral γδ T cell subset, with broad reactivity against stressed human cells, including tumor cells. Vγ9Vδ2 T cells are specifically activated by small phosphorylated metabolites called phosphoantigens (PAg). Stress-induced changes in target cell PAg levels are specifically detected by butyrophilin (BTN)3A1, using its intracellular B30.2 domain. This leads to the activation of Vγ9Vδ2 T cells. In this study, we show that changes in the juxtamembrane domain of BTN3A1, but not its transmembrane domain, induce a markedly enhanced or reduced γδ T cell reactivity. There is thus a specific requirement for BTN3A1's juxtamembrane domain for correct γδ T cell-related function. This work identified, as being of particular importance, a juxtamembrane domain region of BTN3A molecules identified as a possible dimerization interface and that is located close to the start of the B30.2 domain.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/immunology , Butyrophilins/chemistry , Butyrophilins/immunology , Lymphocyte Activation , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Antigens/chemistry , Antigens/immunology , Antigens, CD/metabolism , Butyrophilins/metabolism , HEK293 Cells , Humans , Mutant Chimeric Proteins/immunology , Phosphorylation
10.
Eur J Immunol ; 47(6): 982-992, 2017 06.
Article in English | MEDLINE | ID: mdl-28386905

ABSTRACT

Phosphoantigens (PAgs)-like HMBPP ((E)-4-hydroxy-3-methyl-but-2-enyl diphosphate) and butyrophilin 3 (BTN3A, CD277)-specific monoclonal antibody 20.1 induce TCR-mediated activation of Vγ9Vδ2 T cells. Here, we compared murine reporter cells transduced with Vγ9Vδ2 TCRs G115, D1C55, and MOP for the activation in culture with human RAJI cells and PAgs or mAb 20.1 and its single-chain (sc) derivative. All transductants responded readily to PAg but only TCR MOP γ-chain-expressing cells responded to mAb/sc 20.1. Furthermore, both antagonist and agonist mAb and sc of the agonist mAb inhibited the PAg response of TCR-transduced murine reporter cells. These findings suggest that, in contrast to stimulation by physiological stimulators (PAg), the responsiveness to mAb 20.1 depends strongly on CDR3 sequences of the TCR, and that mAb 20.1 can interfere with the PAg-response. Mouse or human origin of reporter cells might affect the mAb 20.1 response since all three TCR-mediated mAb 20.1-induced activation of TCR-transduced Jurkat cells. The pronounced differences between PAg and mAb 20.1-induced activation observed here help to understand the often contradictory published data. This study provides novel perspectives on the physiological mechanism of Vγ9Vδ2 T-cell activation, and highlights the complex mode of action of BTN3A-specific antibodies as agents in cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Butyrophilins/immunology , Lymphocyte Activation , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Cell Line , HEK293 Cells , Humans , Mice
11.
Immunol Cell Biol ; 95(7): 620-629, 2017 08.
Article in English | MEDLINE | ID: mdl-28356569

ABSTRACT

The inherent resistance of cancer stem cells (CSCs) to existing therapies has largely hampered the development of effective treatments for advanced malignancy. To help develop novel immunotherapy approaches that efficiently target CSCs, an experimental model allowing reliable distinction of CSCs and non-CSCs was set up to study their interaction with non-MHC-restricted γδ T cells and antigen-specific CD8+ T cells. Stable lines with characteristics of breast CSC-like cells were generated from ras-transformed human mammary epithelial (HMLER) cells as confirmed by their CD44hi CD24lo GD2+ phenotype, their mesenchymal morphology in culture and their capacity to form mammospheres under non-adherent conditions, as well as their potent tumorigenicity, self-renewal and differentiation in xenografted mice. The resistance of CSC-like cells to γδ T cells could be overcome by inhibition of farnesyl pyrophosphate synthase (FPPS) through pretreatment with zoledronate or with FPPS-targeting short hairpin RNA. γδ T cells induced upregulation of MHC class I and CD54/ICAM-1 on CSC-like cells and thereby increased the susceptibility to antigen-specific killing by CD8+ T cells. Alternatively, γδ T-cell responses could be specifically directed against CSC-like cells using the humanised anti-GD2 monoclonal antibody hu14.18K322A. Our findings identify a powerful synergism between MHC-restricted and non-MHC-restricted T cells in the eradication of cancer cells including breast CSCs. Our research suggests that novel immunotherapies may benefit from a two-pronged approach combining γδ T-cell and CD8+ T-cell targeting strategies that triggers effective innate-like and tumour-specific adaptive responses.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Animals , Antibodies/pharmacology , Breast/pathology , Cytotoxicity, Immunologic , Diphosphonates/pharmacology , Epithelial Cells/metabolism , Epitopes/immunology , Female , Humans , Imidazoles/pharmacology , Immunity, Innate , Interferon-gamma/metabolism , Major Histocompatibility Complex , Mice , Phenotype , Zoledronic Acid , ras Proteins/metabolism
12.
Clin Immunol ; 158(1): 92-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25796193

ABSTRACT

CD1d-restricted invariant natural killer T (iNKT) cells constitute an important immunoregulatory T cell subset that can be activated by the synthetic glycolipid α-galactosylceramide (α-GalCer) and initiate antitumor immune responses. As cancer patients are frequently treated with aminobisphosphonates (NBP), it is relevant to determine possible effects of NBP on CD1d-restricted glycolipid Ag-presentation to iNKT cells. We report a striking reduction of α-GalCer-induced iNKT cell activation by monocyte derived dendritic cells (moDC) upon their exposure to NBP during maturation. We found that production of apolipoprotein E (apoE), which is a known facilitator of trans-membrane transport of exogenously derived glycolipids, was significantly diminished in moDC exposed to NBP. As the inhibitory effect of NBP on iNKT cell activation was alleviated by exogenous apoE, our data indicate that reduced apoE production by antigen presenting cells (APC) through NBP limits glycolipid-induced iNKT cell activation. This should be taken into account in the design of iNKT cell-based anti-cancer therapies.


Subject(s)
Amines/pharmacology , Bone Density Conservation Agents/pharmacology , Dendritic Cells/drug effects , Diphosphonates/pharmacology , Lymphocyte Activation/drug effects , Natural Killer T-Cells/drug effects , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigens, CD1d/immunology , Cell Line , Dendritic Cells/immunology , Galactosylceramides/pharmacology , Humans , Natural Killer T-Cells/immunology
13.
J Immunol ; 191(4): 1993-2000, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23836057

ABSTRACT

Peripheral Vγ9Vδ2 T lymphocytes compose a major γδ T cell subset in primates with broad reactivity against tumor cells. Vγ9Vδ2 T cells are specifically activated by phosphorylated isoprenoid pathway metabolites called "phosphoagonists." Accordingly, pharmacologic inhibitors of the mevalonate pathway, such as aminobisphosphonates (NBP) that upregulate the intracellular production of phosphoagonists, increase antitumor Vγ9Vδ2 T cell responses. Immunotherapeutic protocols exploiting GMP-grade agonist molecules targeting human Vγ9Vδ2 T lymphocytes have yielded promising, yet limited, signs of antitumor efficacy and therefore need to be improved for next-generation immunotherapies. In this study, we used a model of s.c. human tumor xenografts in severely immunodeficient mice to assess the antitumor efficacy of systemic NBP treatments when combined with the adoptive transfer of human Vγ9Vδ2 T cells. We show that infusion of Vγ9Vδ2 T cells, 24 h after systemic NBP treatment, efficiently delays tumor growth in mice. Importantly, our results indicate efficient but transient in vivo NBP-induced sensitization of tumor cells to human Vγ9Vδ2-T cell recognition. Accordingly, repeated and combined administrations of both NBP and γδ T cells yielded improved antitumor responses in vivo. Because Vγ9Vδ2 T cells show similar responsiveness toward both autologous and allogeneic tumors and are devoid of alloreactivity, these results provide preclinical proof of concept for optimized antitumor immunotherapies combining NBP treatment and adoptive transfer of allogeneic human γδ T cells.


Subject(s)
Adenocarcinoma/therapy , Diphosphonates/therapeutic use , Immunotherapy, Adoptive , Prostatic Neoplasms/therapy , T-Lymphocyte Subsets/transplantation , Adenocarcinoma/pathology , Animals , Cell Line, Tumor/transplantation , Diphosphonates/administration & dosage , Drug Administration Schedule , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lymphocyte Activation , Male , Mevalonic Acid/metabolism , Mice , Mice, Mutant Strains , Mice, SCID , Pamidronate , Prostatic Neoplasms/pathology , Receptors, Antigen, T-Cell, gamma-delta/analysis , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/immunology , Xenograft Model Antitumor Assays
14.
J Immunol ; 191(1): 30-4, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23740951

ABSTRACT

Human γδ T cells expressing the Vδ3 TCR make up a minor lymphocyte subset in blood but are enriched in liver and in patients with some chronic viral infections and leukemias. We analyzed the frequencies, phenotypes, restriction elements, and functions of fresh and expanded peripheral blood Vδ3 T cells. Vδ3 T cells accounted for ~0.2% of circulating T cells, included CD4(+), CD8(+), and CD4(-)CD8(-) subsets, and variably expressed CD56, CD161, HLA-DR, and NKG2D but neither NKG2A nor NKG2C. Vδ3 T cells were sorted and expanded by mitogen stimulation in the presence of IL-2. Expanded Vδ3 T cells recognized CD1d but not CD1a, CD1b, or CD1c. Upon activation, they killed CD1d(+) target cells, released Th1, Th2, and Th17 cytokines, and induced maturation of dendritic cells into APCs. Thus, Vδ3 T cells are glycolipid-reactive T cells with distinct Ag specificities but functional similarities to NKT cells.


Subject(s)
Antigens, CD1d/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Antigens, CD1d/metabolism , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunophenotyping
15.
Blood ; 120(11): 2269-79, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22767497

ABSTRACT

Human peripheral Vγ9Vδ2 T cells are activated by phosphorylated metabolites (phosphoagonists [PAg]) of the mammalian mevalonate or the microbial desoxyxylulose-phosphate pathways accumulated by infected or metabolically distressed cells. The underlying mechanisms are unknown. We show that treatment of nonsusceptible target cells with antibody 20.1 against CD277, a member of the extended B7 superfamily related to butyrophilin, mimics PAg-induced Vγ9Vδ2 T-cell activation and that the Vγ9Vδ2 T-cell receptor is implicated in this effect. Vγ9Vδ2 T-cell activation can be abrogated by exposing susceptible cells (tumor and mycobacteria-infected cells, or aminobisphosphonate-treated cells with up-regulated PAg levels) to antibody 103.2 against CD277. CD277 knockdown and domain-shuffling approaches confirm the key implication of the CD277 isoform BTN3A1 in PAg sensing by Vγ9Vδ2 T cells. Fluorescence recovery after photobleaching (FRAP) experiments support a causal link between intracellular PAg accumulation, decreased BTN3A1 membrane mobility, and ensuing Vγ9Vδ2 T-cell activation. This study demonstrates a novel role played by B7-like molecules in human γδ T-cell antigenic activation and paves the way for new strategies to improve the efficiency of immunotherapies using Vγ9Vδ2 T cells.


Subject(s)
Antigens, CD/metabolism , Antigens/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/metabolism , Antibodies, Blocking , Antibodies, Immobilized , Antibodies, Monoclonal , Antigens, CD/chemistry , Antigens, CD/genetics , Butyrophilins , Cells, Cultured , Clone Cells , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Immunologic Factors/pharmacology , Lymphocyte Activation/drug effects , Phosphorylation/drug effects , Protein Isoforms/agonists , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , RNA, Small Interfering , Receptors, Antigen, T-Cell/agonists , Receptors, Antigen, T-Cell/antagonists & inhibitors , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
16.
Semin Immunol ; 22(4): 199-206, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20447835

ABSTRACT

Human Vgamma9Vdelta2 T cells, a major innate-like peripheral T cell subset, are thought to play in vivo a key role in innate and adaptive immune responses to infection agents and tumors. Vgamma9Vdelta2 T cell activation is tightly regulated by a variety of activating or inhibitory receptors which are specific for constitutively expressed or stress-modulated ligands. However, the mechanisms and signal transduction pathways regulating their broad effector functions, such as cytotoxicity and cytokine responses, remain poorly understood. Here we provide an updated overview of the activation modalities of Vgamma9Vdelta2 T cells by highlighting the respective role played by T cell receptor (TCR) versus non-TCR stimuli, and focus on recent studies showing how Vgamma9Vdelta2 T cells integrate the numerous activating and inhibitory signals and translate them into a particular effector and biological function. A better understanding of these critical issues should help optimize immunotherapeutic approaches targeting Vgamma9Vdelta2 T cells.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta/immunology , Signal Transduction , T-Lymphocytes/immunology , Animals , Humans , Lymphocyte Activation , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Natural Killer Cell/immunology , T-Lymphocytes/metabolism
17.
J Biol Chem ; 287(39): 32780-90, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22846996

ABSTRACT

Human Vγ9Vδ2 T cells are well known for their rapid and potent response to infection and tumorigenesis when in the presence of endogenous or exogenous phosphoisoprenoids. However, the molecular mechanisms behind the activation of this γδ T cell population remains unclear. Evidence pointing to a role for the CD277/butyrophilin-3 (BTN3A) molecules in this response led us to investigate the structures of these molecules and their modifications upon binding to an agonist antibody (20.1) that mimics phosphoisoprenoid-mediated Vγ9Vδ2 activation and an antagonist antibody (103.2) that inhibits this reactivity. We find that the three BTN3A isoforms: BTN3A1, BTN3A2, and BTN3A3, have high structural homology to the B7 superfamily of proteins and exist as V-shaped homodimers in solution, associating through the membrane proximal C-type Ig domain. The 20.1 and 103.2 antibodies bind to separate epitopes on the BTN3A Ig-V domain with high affinity but likely with different valencies based on their binding orientation. These structures directly complement functional studies of this system that demonstrate that BTN3A1 is necessary for Vγ9Vδ2 activation and begin to unravel the extracellular events that occur during stimulation through the Vγ9Vδ2 T cell receptor.


Subject(s)
Antibodies/immunology , Antigens, CD/immunology , Lymphocyte Activation , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Antibodies/chemistry , Antigens, CD/chemistry , Antigens, CD/genetics , Butyrophilins , Humans , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, gamma-delta/chemistry , Receptors, Antigen, T-Cell, gamma-delta/genetics , Structural Homology, Protein , Structure-Activity Relationship , T-Lymphocytes/chemistry
18.
Blood ; 117(10): 2864-73, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21233315

ABSTRACT

In humans, the majority of peripheral blood γδ T cells expresses Vγ9Vδ2 T-cell receptors (TCR) and recognize nonpeptidic phosphorylated antigens. In contrast, most tissue-derived γδ T cells, which are located mainly in spleen and epithelia, preferentially use Vδ1 or Vδ3 chains paired with diverse Vγ chains to form their TCR. Our knowledge about the antigenic specificity and costimulation requirements of human Vδ2(-) γδ T cells remains limited. In an attempt to address this important issue, we characterized the specificity of a monoclonal antibody (mAb 256), screened for its ability to specifically inhibit cytolytic responses of several human Vδ2(-) γδ T-cell clones against transformed B cells. We show that mAb 256 does not target a TCR ligand but blocks key interactions between non-TCR molecules on effector γδ T cells and ILT2 molecule, expressed by tumor targets. In line with the previously reported specificity of this NK receptor for classic and nonclassic major histocompatibility complex (MHC) class I molecules, blockade of MHC class I/ILT2 interactions using MHC class I- or ILT2-specific mAbs and ILT2-Fc molecules inhibited tumor-induced activation of Vγ8Vδ3 T-cell clones. Therefore, this study describes a new cytotoxic T lymphocyte activation pathway involving MHC class I engagement on γδ T cells.


Subject(s)
Antigens, CD/immunology , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, CD/metabolism , Blotting, Western , Histocompatibility Antigens Class I/metabolism , Humans , Leukocyte Immunoglobulin-like Receptor B1 , Microscopy, Confocal , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Immunologic/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Up-Regulation
19.
Clin Immunol ; 142(2): 194-200, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22122798

ABSTRACT

Vγ9Vδ2-T cells constitute a proinflammatory lymphocyte subpopulation with established antitumor activity. Phosphoantigens activate Vγ9Vδ2-T cells in vivo and in vitro. We studied whether the antitumor activity of Vγ9Vδ2-T cells can be potentiated by invariant NKT cells (iNKT), an important immunoregulatory T cell subset. When activated by the glycolipid α-galactosylceramide (α-GalCer), iNKT produce large amounts of cytokines involved in antitumor immune responses. Monocyte-derived dendritic cells were loaded with both phosphoantigens (using aminobisphosphonates) and α-GalCer during maturation and subsequently co-cultured with Vγ9Vδ2-T and iNKT cells. Aminobisphosphonates dose-dependently enhanced Vγ9Vδ2-T cell activation, and this was potentiated by α-GalCer-induced iNKT co-activation. iNKT co-activation also enhanced the IFN-γ production and cytolytic potential of Vγ9Vδ2-T cells against tumor cells. Using transwell experiments and neutralizing antibodies cross-talk between iNKT and Vγ9Vδ2-T cells was found to be mediated by TNF-α. Our data provide a rationale for combining both activating ligands to improve Vγ9Vδ2-T cell based approaches in cancer-immunotherapy.


Subject(s)
Lymphocyte Activation/immunology , Natural Killer T-Cells , Neoplasms , T-Lymphocytes, Cytotoxic , Tumor Necrosis Factor-alpha/biosynthesis , Antigens, Neoplasm/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diphosphonates/pharmacology , Galactosylceramides/immunology , Galactosylceramides/pharmacology , Hemiterpenes/metabolism , Humans , Immunologic Factors/immunology , Immunotherapy/methods , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Neoplasms/immunology , Neoplasms/therapy , Organophosphorus Compounds/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Necrosis Factor-alpha/immunology
20.
J Immunol ; 185(1): 55-63, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20511557

ABSTRACT

Human Vgamma9Vdelta2 T cells, a major innate-like peripheral T cell subset, are thought to play in vivo an important role in innate and adaptive immune responses to infection agents and tumors. However, the mechanisms regulating their broad effector functions, such as cytotoxicity and cytokine responses, remain poorly understood. In this study, we used single-cell calcium video imaging to analyze the early intracellular events associated with TCR-induced Vgamma9Vdelta2 T cell functional responses. When compared with other human T cell subsets, including NKT and Vdelta2(neg) gammadelta T cells, TCR/CD3-activated Vgamma9Vdelta2 T cells displayed an unusually delayed and sustained intracellular calcium mobilization, which was dramatically quickened and shortened on costimulation by NKG2D, a main activating NKR regulating gammadelta T cell tumor cytolysis. Importantly, the protein kinase C transduction pathway was identified as a main regulator of the NKG2D-mediated costimulation of antitumor Vgamma9Vdelta2 cytolytic responses. Therefore, this study identifies a new mechanism regulating Vgamma9Vdelta2 T cell functional plasticity through fine-tuning of early signal transduction events.


Subject(s)
Calcium Signaling/immunology , Cytotoxicity Tests, Immunologic , Isoenzymes/physiology , NK Cell Lectin-Like Receptor Subfamily K/physiology , Natural Killer T-Cells/immunology , Neoplasms, Experimental/immunology , Protein Kinase C/physiology , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Receptors, Antigen, T-Cell/physiology , Animals , CD3 Complex/biosynthesis , CD3 Complex/physiology , Cell Communication/immunology , Cell Line, Tumor , Clone Cells , Cytotoxicity Tests, Immunologic/methods , Enzyme Induction/immunology , Humans , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lymphocyte Activation/immunology , MAP Kinase Signaling System/immunology , Mice , Natural Killer T-Cells/enzymology , Natural Killer T-Cells/metabolism , Neoplasms, Experimental/prevention & control , Protein Kinase C-theta , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL