Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1426620, 2024.
Article in English | MEDLINE | ID: mdl-39035010

ABSTRACT

The nucleotide-binding and oligomerization domain-like receptors (NLRs) NLR family CARD domain-containing protein 5 (NLRC5) and Class II Major Histocompatibility Complex Transactivator (CIITA) are transcriptional regulators of major histocompatibility complex (MHC) class I and class II genes, respectively. MHC molecules are central players in our immune system, allowing the detection of hazardous 'non-self' antigens and, thus, the recognition and elimination of infected or transformed cells from the organism. Recently, CIITA and NLRC5 have emerged as regulators of selected genes of the butyrophilin (BTN) family that interestingly are located in the extended MHC locus. BTNs are transmembrane proteins exhibiting structural similarities to B7 family co-modulatory molecules. The family member BTN2A2, which indeed contributes to the control of T cell activation, was found to be transcriptionally regulated by CIITA. NLRC5 emerged instead as an important regulator of the BTN3A1, BTN3A2, and BTN3A3 genes. Together with BTN2A1, BTN3As regulate non-conventional Vγ9Vδ2 T cell responses triggered by selected metabolites of microbial origin or accumulating in hematologic cancer cells. Even if endogenous metabolites conform to the canonical definition of 'self', metabolically abnormal cells can represent a danger for the organism and should be recognized and controlled by immune system cells. Collectively, new data on the role of NLRC5 in the expression of BTN3As link the mechanisms regulating canonical 'non-self' presentation and those marking cells with abnormal metabolic configurations for immune recognition, an evolutionary parallel that we discuss in this perspective review.


Subject(s)
Butyrophilins , Intracellular Signaling Peptides and Proteins , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Butyrophilins/metabolism , Butyrophilins/genetics , Butyrophilins/immunology , Animals , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Gene Expression Regulation , Lymphocyte Activation/immunology , Antigens, CD
2.
Eur J Clin Invest ; 54(8): e14207, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38558028

ABSTRACT

BACKGROUND: Cytometric analysis has been commonly used to delineate distinct cell subpopulations among peripheral blood mononuclear cells by the differential expression of surface receptors. This capability has reached its apogee with high-dimensional approaches such as mass cytometry and spectral cytometry that include simultaneous assessment of 20-50 analytes. Unfortunately, this approach also engenders significant complexity with analytical and interpretational pitfalls. METHODS: Here, we demonstrate a complementary approach with restricted-dimensionality to assess cell-type specific intracellular molecular expression levels at exceptional levels of precision. The expression of five analytes was individually assessed in four mononuclear cell-types from peripheral blood. RESULTS: Distinctions in expression levels were seen between cell-types and between samples from different donor groups. Mononuclear cell-type specific molecular expression levels distinguished pregnant from nonpregnant women and G-CSF-treated from untreated persons. Additionally, the precision of our analysis was sufficient to quantify a novel relationship between two molecules-Rel A and translocator protein-by correlational analysis. CONCLUSIONS: Restricted-dimensional cytometry can provide a complementary approach to define characteristics of cell-type specific intracellular protein and phosphoantigen expression in mononuclear cells.


Subject(s)
Flow Cytometry , Leukocytes, Mononuclear , Humans , Female , Leukocytes, Mononuclear/metabolism , Pregnancy , Adult , Male
3.
Res Sq ; 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36824912

ABSTRACT

Butyrophilin (BTN)-3A and BTN2A1 molecules control TCR-mediated activation of human Vγ9Vδ2 T-cells triggered by phosphoantigens (PAg) from microbes and tumors, but the molecular rules governing antigen sensing are unknown. Here we establish three mechanistic principles of PAg-action. Firstly, in humans, following PAg binding to the BTN3A1-B30.2 domain, Vγ9Vδ2 TCR triggering involves the V-domain of BTN3A2/BTN3A3. Moreover, PAg/B30.2 interaction, and the critical γδ-T-cell-activating V-domain, localize to different molecules. Secondly, this distinct topology as well as intracellular trafficking and conformation of BTN3A heteromers or ancestral-like BTN3A homomers are controlled by molecular interactions of the BTN3 juxtamembrane region. Finally, the ability of PAg not simply to bind BTN3A-B30.2, but to promote its subsequent interaction with the BTN2A1-B30.2 domain, is essential for T-cell activation. Defining these determinants of cooperation and division of labor in BTN proteins deepens understanding of PAg sensing and elucidates a mode of action potentially applicable to other BTN/BTNL family members.

6.
Front Immunol ; 13: 915837, 2022.
Article in English | MEDLINE | ID: mdl-35784326

ABSTRACT

γδ T-cells directly recognize and kill transformed cells independently of HLA-antigen presentation, which makes them a highly promising effector cell compartment for cancer immunotherapy. Novel γδ T-cell-based immunotherapies, primarily focusing on the two major γδ T-cell subtypes that infiltrate tumors (i.e. Vδ1 and Vδ2), are being developed. The Vδ1 T-cell subset is enriched in tissues and contains both effector T-cells as well as regulatory T-cells with tumor-promoting potential. Vδ2 T-cells, in contrast, are enriched in circulation and consist of a large, relatively homogeneous, pro-inflammatory effector T-cell subset. Healthy individuals typically harbor in the order of 50-500 million Vγ9Vδ2 T-cells in the peripheral blood alone (1-10% of the total CD3+ T-cell population), which can rapidly expand upon stimulation. The Vγ9Vδ2 T-cell receptor senses intracellular phosphorylated metabolites, which accumulate in cancer cells as a result of mevalonate pathway dysregulation or upon pharmaceutical intervention. Early clinical studies investigating the therapeutic potential of Vγ9Vδ2 T-cells were based on either ex vivo expansion and adoptive transfer or their systemic activation with aminobisphosphonates or synthetic phosphoantigens, either alone or combined with low dose IL-2. Immune-related adverse events (irAE) were generally \mild, but the clinical efficacy of these approaches provided overall limited benefit. In recent years, critical advances have renewed the excitement for the potential of Vγ9Vδ2 T-cells in cancer immunotherapy. Here, we review γδ T-cell-based therapeutic strategies and discuss the prospects of those currently evaluated in clinical studies in cancer patients as well as future therapies that might arise from current promising pre-clinical results.


Subject(s)
Intraepithelial Lymphocytes , Neoplasms , Humans , Immunotherapy , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets
7.
Biol Open ; 11(1)2022 02 15.
Article in English | MEDLINE | ID: mdl-34994391

ABSTRACT

Vγ9Vδ2 T cells is the dominant γδ T cell subset in human blood. They are cytotoxic and activated by phosphoantigens whose concentrations are increased in cancer cells, making the cancer cells targets for Vγ9Vδ2 T cell immunotherapy. For successful immunotherapy, it is important both to characterise Vγ9Vδ2 T cell proliferation and optimise the assessment of their cytotoxic potential, which is the aim of this study. We found that supplementation with freshly thawed human serum potentiated Vγ9Vδ2 T cell proliferation from peripheral mononuclear cells (PBMCs) stimulated with (E)-4-Hydroxy-3-methyl-but-2-enyl diphosphate (HMBPP) and consistently enabled Vγ9Vδ2 T cell proliferation from cryopreserved PBMCs. In cryopreserved PBMCs the proliferation was higher than in freshly prepared PBMCs. In a panel of short-chain prenyl alcohols, monophosphates and diphosphates, most diphosphates and also dimethylallyl monophosphate stimulated Vγ9Vδ2 T cell proliferation. We developed a method where the cytotoxicity of Vγ9Vδ2 T cells towards adherent cells is assessed at the single cell level using flow cytometry, which gives more clear-cut results than the traditional bulk release assays. Moreover, we found that HMBPP enhances the Vγ9Vδ2 T cell cytotoxicity towards colon cancer cells. In summary, we have developed an easily interpretable method to assess the cytotoxicity of Vγ9Vδ2 T cells towards adherent cells, found that Vγ9Vδ2 T cell proliferation can be potentiated by media-supplementation and how misclassification of non-responders may be avoided. Our findings will be useful in the further development of Vγ9Vδ2 T cell immunotherapy.


Subject(s)
Neoplasms , Receptors, Antigen, T-Cell, gamma-delta , Cell Proliferation , Flow Cytometry , Humans , Lymphocyte Activation , Neoplasms/therapy , T-Lymphocytes
8.
Adv Exp Med Biol ; 1273: 91-104, 2020.
Article in English | MEDLINE | ID: mdl-33119877

ABSTRACT

Gamma delta (γδ) T cells which combine both innate and adaptive potential have extraordinary properties. Indeed, their strong cytotoxic and pro-inflammatory activity allows them to kill a broad range of tumor cells. Several studies have demonstrated that γδ T cells are an important component of tumor-infiltrated lymphocytes in patients affected by different types of cancer. Tumor-infiltrating γδ T cells are also considered as a good prognostic marker in many studies, though the presence of these cells is associated with poor prognosis in breast and colon cancers. The tumor microenvironment seems to drive γδ T-cell differentiation toward a tumor-promoting or a tumor-controlling phenotype, which suggests that some tumor microenvironments can limit the effectiveness of γδ T cells.The major γδ T-cell subsets in human are the Vγ9Vδ2 T cells that are specifically activated by phosphoantigens. This unique antigenic activation process operates in a framework that requires the expression of butyrophilin 3A (BTN3A) molecules. Interestingly, there is some evidence that BTN3A expression may be regulated by the tumor microenvironment. Given their strong antitumoral potential, Vγ9Vδ2 T cells are used in therapeutic approaches either by ex vivo culture and amplification, and then adoptive transfer to patients or by direct stimulation to propagate in vivo. These strategies have demonstrated promising initial results, but greater potency is needed. Combining Vγ9Vδ2 T-cell immunotherapy with systemic approaches to restore antitumor immune response in tumor microenvironment may improve efficacy.In this chapter, we first review the basic features of γδ T cells and their roles in the tumor microenvironment and then analyze the advances about the understanding of these cells' activation in tumors and why this represent unique challenges for therapeutics, and finally we discuss γδ T-cell-based therapeutic strategies and future perspectives of their development.


Subject(s)
Neoplasms/immunology , T-Lymphocyte Subsets/cytology , Tumor Microenvironment/immunology , Butyrophilins/immunology , Cell Differentiation , Humans , Lymphocyte Activation , Neoplasms/therapy , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets/immunology
9.
Immunol Rev ; 298(1): 74-83, 2020 11.
Article in English | MEDLINE | ID: mdl-33017054

ABSTRACT

Gamma-delta (γδ) T cells are an important component of the immune system. They are often enriched in non-lymphoid tissues and exhibit diverse functional attributes including rapid activation, cytokine production, proliferation, and acquisition of cytotoxicity following both TCR-dependent and TCR-independent stimulation, but poor capacity for immunological memory. They can detect a broad range of antigens, although typically not peptide-MHC complexes in contrast to alpha-beta (αß) T cells. In humans, a prominent population of γδ T cells, defined as Vγ9Vδ2+ cells, reacts to small phosphorylated non-peptide "phosphoantigens" (pAgs). The molecular mechanism underpinning this recognition is poorly defined, but is known to involve butyrophilin family members and appears to involve indirect pAg recognition via alterations to butyrophilin molecular complexes. In this review, we discuss recent advances in our understanding of pAg recognition by γδ T cells including the role of butyrophilins and in particular, a newly described role for butyrophilin 2A1.


Subject(s)
Lymphocyte Activation , Receptors, Antigen, T-Cell, gamma-delta , Antigens, CD , Butyrophilins , Humans , T-Lymphocytes
10.
ChemMedChem ; 14(17): 1597-1603, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31226236

ABSTRACT

A set of phosphonate prodrugs of a butyrophilin ligand was synthesized and evaluated for plasma stability and cellular activity. The mixed aryl acyloxy esters were prepared either via a standard sequence through the phosphonic acid chloride, or through the more recently reported, and more facile, triflate activation. In the best of cases, this class of prodrugs shows cellular potency similar to that of bis-acyloxyalkyl phosphonate prodrugs and plasma stability similar to that of aryl phosphonamidates. For example, {[((3E)-5-hydroxy-4-methylpent-3-en-1-yl) (naphthalen-2-yloxy)phosphoryl]oxy}methyl 2,2-dimethylpropanoate can activate BTN3A1 in K562 cells after just 15 minutes of exposure (at an EC50 value of 31 nm) and is only partially metabolized (60 % remaining) after 20 hours in human plasma. Other related novel analogues showed similar potency/stability profiles. Therefore, mixed aryl acyloxyalkyl phosphonate prodrugs are an exciting new strategy for the delivery of phosphonate-containing drugs.


Subject(s)
Butyrophilins/pharmacology , Organophosphonates/pharmacology , Prodrugs/pharmacology , Butyrophilins/blood , Butyrophilins/chemical synthesis , Butyrophilins/toxicity , Drug Stability , Humans , K562 Cells , Organophosphonates/blood , Organophosphonates/chemical synthesis , Organophosphonates/toxicity , Prodrugs/chemical synthesis , Prodrugs/toxicity
11.
Immunity ; 50(4): 1043-1053.e5, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30902636

ABSTRACT

Human Vγ9Vδ2 T cells respond to microbial infections and malignancy by sensing diphosphate-containing metabolites called phosphoantigens, which bind to the intracellular domain of butyrophilin 3A1, triggering extracellular interactions with the Vγ9Vδ2 T cell receptor (TCR). Here, we examined the molecular basis of this "inside-out" triggering mechanism. Crystal structures of intracellular butyrophilin 3A proteins alone or in complex with the potent microbial phosphoantigen HMBPP or a synthetic analog revealed key features of phosphoantigens and butyrophilins required for γδ T cell activation. Analyses with chemical probes and molecular dynamic simulations demonstrated that dimerized intracellular proteins cooperate in sensing HMBPP to enhance the efficiency of γδ T cell activation. HMBPP binding to butyrophilin doubled the binding force between a γδ T cell and a target cell during "outside" signaling, as measured by single-cell force microscopy. Our findings provide insight into the "inside-out" triggering of Vγ9Vδ2 T cell activation by phosphoantigen-bound butyrophilin, facilitating immunotherapeutic drug design.


Subject(s)
Antigens, CD/chemistry , Butyrophilins/chemistry , Lymphocyte Activation , Organophosphates/metabolism , T-Lymphocyte Subsets/immunology , Antigens, CD/metabolism , Binding Sites , Butyrophilins/metabolism , Crystallography, X-Ray , Dimerization , Drug Design , Humans , Hydrogen Bonding , Immunotherapy , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Conformation , Protein Domains , Protein Isoforms/chemistry , Protein Processing, Post-Translational , Receptors, Antigen, T-Cell, gamma-delta , Single-Cell Analysis , Structure-Activity Relationship , T-Lymphocyte Subsets/metabolism
13.
Front Immunol ; 9: 1246, 2018.
Article in English | MEDLINE | ID: mdl-29937767

ABSTRACT

Human Vγ9Vδ2 T cells have the capacity to detect supra-physiological concentrations of phosphoantigens (pAgs) generated by the mevalonate (Mev) pathway of mammalian cells under specific circumstances. Isopentenyl pyrophosphate (IPP) is the prototypic pAg recognized by Vγ9Vδ2 T cells. B-cell derived tumor cells (i.e., lymphoma and myeloma cells) and dendritic cells (DCs) are privileged targets of Vγ9Vδ2 T cells because they generate significant amounts of IPP which can be boosted with zoledronic acid (ZA). ZA is the most potent aminobisphosphonate (NBP) clinically available to inhibit osteoclast activation and a very potent inhibitor of farnesyl pyrophosphate synthase in the Mev pathway. ZA-treated DCs generate and release in the supernatants picomolar IPP concentrations which are sufficient to induce the activation of Vγ9Vδ2 T cells. We have recently shown that the ATP-binding cassette transporter A1 (ABCA1) plays a major role in the extracellular release of IPP from ZA-treated DCs. This novel ABCA1 function is fine-tuned by physical interactions with IPP, apolipoprotein A-I (apoA-I), and butyrophilin-3A1 (BTN3A1). The mechanisms by which soluble IPP induces Vγ9Vδ2 T-cell activation remain to be elucidated. It is possible that soluble IPP binds to BTN3A1, apoA-I, or other unknown molecules on the cell surface of bystander cells like monocytes, NK cells, Vγ9Vδ2 T cells, or any other cell locally present. Investigating this scenario may represent a unique opportunity to further characterize the role of BTN3A1 and other molecules in the recognition of soluble IPP by Vγ9Vδ2 T cells.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Butyrophilins/genetics , Butyrophilins/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Antigens/immunology , Antigens/metabolism , Cell Membrane/metabolism , Gene Expression Regulation , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Phosphorylation , Protein Binding , Protein Transport , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
14.
Front Immunol ; 9: 828, 2018.
Article in English | MEDLINE | ID: mdl-29731756

ABSTRACT

Vγ9Vδ2 T cells represent a major unconventional γδ T cell subset located in the peripheral blood of adults in humans and several non-human primates. Lymphocytes that constitute this transitional subset can sense subtle level changes of intracellular phosphorylated intermediates of the isoprenoid biosynthesis pathway (phosphoantigens, pAg), such as isopentenyl pyrophosphate, during cell stress events. This unique antigenic activation process operates in a rigorous framework that requires the expression of butyrophilin 3A1 (BTN3A1/CD277) molecules, which are type I glycoproteins that belong to the B7 family. Several studies have further shown that pAg specifically bind to the intracellular B30.2 domain of BTN3A1 linked to the antigenic activation of Vγ9Vδ2 T cells. Here, we highlight the recent advances in BTN3A1 dynamics induced upon the binding of pAg and the contribution of the different subunits to this activation process. Recent reports support that conformational modifications of BTN3A1 might represent a key step in the detection of infection or tumorigenesis by Vγ9Vδ2 T cells. A better understanding of this mechanism will help optimize novel immunotherapeutical approaches that target defined functions of this unique γδ T cell subset.


Subject(s)
Antigens, CD/immunology , Antigens/immunology , Butyrophilins/immunology , Lymphocyte Activation , T-Lymphocyte Subsets/immunology , Amino Acid Sequence , HEK293 Cells , Humans , Phosphorylation , Protein Binding , Receptors, Antigen, T-Cell, gamma-delta
15.
Front Immunol ; 9: 753, 2018.
Article in English | MEDLINE | ID: mdl-29706966

ABSTRACT

Objectives: γδ T cells, a non-conventional innate lymphocyte subset containing cells that can be activated by lipids and phosphoantigens, are abnormally regulated in systemic sclerosis (SSc). To further evaluate the significance of this dysregulation, we compared how exposure to an autoantigenic lipid, cardiolipin (CL), during co-stimulation with an amino-bisphosphonate (zoledronate, zol), affects the activation and cytokine production of SSc and healthy control (HC) γδ T cells. Methods: Expression of CD25 on Vγ9+, Vδ1+, and total CD3+ T cells in cultured peripheral blood mononuclear cells (PBMCs), their binding of CD1d tetramers, and the effect of monoclonal antibody (mAb) blockade of CD1d were monitored by flow cytometry after 4 days of in vitro culture. Intracellular production of IFNγ and IL-4 was assessed after overnight culture. Results: Percentages of CD25+ among CD3+ and Vδ1+ T cells were elevated significantly in short-term cultured SSc PBMC compared to HC. In SSc but not HC, CL and zol, respectively, suppressed %CD25+ Vγ9+ and Vδ1+ T cells but, when combined, CL + zol significantly activated both subsets in HC and partially reversed inhibition by the individual reagents in SSc. Importantly, Vδ1+ T cells in both SSc and HC were highly reactive with lipid presenting CD1d tetramers, and a CD1d-blocking mAb decreased CL-induced enhancement of %SSc CD25+ Vδ1+ T cells in the presence of zol. %IFNγ+ cells among Vγ9+ T cells of SSc was lower than HC cultured in medium, CL, zol, or CL + zol, whereas %IFNγ+ Vδ1+ T cells was lower only in the presence of CL or CL + zol. %IL-4+ T cells were similar in SSc and HC in all conditions, with the exception of being increased in SSc Vγ9+ T cells in the presence of CL. Conclusion: Abnormal functional responses of γδ T cell subsets to stimulation by CL and phosphoantigens in SSc may contribute to fibrosis and immunosuppression, characteristics of this disease.


Subject(s)
Cardiolipins/pharmacology , Lymphocyte Activation/immunology , Scleroderma, Systemic/immunology , T-Lymphocyte Subsets/immunology , Zoledronic Acid/pharmacology , Adult , Aged , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Diphosphonates/pharmacology , Female , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation/drug effects , Male , Middle Aged , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets/drug effects
16.
Front Immunol ; 9: 662, 2018.
Article in English | MEDLINE | ID: mdl-29670629

ABSTRACT

Activation of human Vγ9/Vδ2 T cells by "phosphoantigens" (pAg), the microbial metabolite (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMB-PP) and the endogenous isoprenoid intermediate isopentenyl pyrophosphate, requires expression of butyrophilin BTN3A molecules by presenting cells. However, the precise mechanism of activation of Vγ9/Vδ2 T cells by BTN3A molecules remains elusive. It is not clear what conformation of the three BTN3A isoforms transmits activation signals nor how externally delivered pAg accesses the cytosolic B30.2 domain of BTN3A1. To approach these problems, we studied two HLA haplo-identical HeLa cell lines, termed HeLa-L and HeLa-M, which showed marked differences in pAg-dependent stimulation of Vγ9/Vδ2 T cells. Levels of IFN-γ secretion by Vγ9/Vδ2 T cells were profoundly increased by pAg loading, or by binding of the pan-BTN3A specific agonist antibody CD277 20.1, in HeLa-M compared to HeLa-L cells. IL-2 production from a murine hybridoma T cell line expressing human Vγ9/Vδ2 T cell receptor (TCR) transgenes confirmed that the differential responsiveness to HeLa-L and HeLa-M was TCR dependent. By tissue typing, both HeLa lines were shown to be genetically identical and full-length transcripts of the three BTN3A isoforms were detected in equal abundance with no sequence variation. Expression of BTN3A and interacting molecules, such as periplakin or RhoB, did not account for the functional variation between HeLa-L and HeLa-M cells. Instead, the data implicate a checkpoint controlling BTN3A1 stability and protein trafficking, acting at an early time point in its maturation. In addition, plasma membrane profiling was used to identify proteins upregulated in HMB-PP-treated HeLa-M. ABCG2, a member of the ATP-binding cassette (ABC) transporter family was the most significant candidate, which crucially showed reduced expression in HeLa-L. Expression of a subset of ABC transporters, including ABCA1 and ABCG1, correlated with efficiency of T cell activation by cytokine secretion, although direct evidence of a functional role was not obtained by knockdown experiments. Our findings indicate a link between members of the ABC protein superfamily and the BTN3A-dependent activation of γδ T cells by endogenous and exogenous pAg.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Antigens, CD/metabolism , Butyrophilins/metabolism , Neoplasm Proteins/metabolism , T-Lymphocytes/immunology , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Antigen Presentation , Antigens, CD/genetics , Butyrophilins/genetics , Diphosphates/immunology , HLA Antigens/metabolism , HeLa Cells , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Protein Stability , Receptors, Antigen, T-Cell, gamma-delta/metabolism
17.
Semin Cell Dev Biol ; 84: 65-74, 2018 12.
Article in English | MEDLINE | ID: mdl-29471037

ABSTRACT

Despite playing critical roles in the immune response and having significant potential in immunotherapy, γδ T cells have garnered little of the limelight. One major reason for this paradox is that their antigen recognition mechanisms are largely unknown, limiting our understanding of their biology and our potential to modulate their activity. One of the best-studied γδ subsets is the human Vγ9Vδ2T cell population, which predominates in peripheral blood and can combat both microbial infections and cancers. Although it has been known for decades that Vγ9Vδ2T cells respond to the presence of small pyrophosphate-based metabolites, collectively named phosphoantigens (pAgs), derived from microbial sources or malignant cells, the molecular basis for this response has been unclear. A major breakthrough in this area came with the identification of the Butyrophilin 3A (BTN3A) proteins, members of the Butyrophilin/Butyrophilin-like protein family, as mediators between pAgs and Vγ9Vδ2T cells. In this article, we review the most recent studies regarding pAg activation of human Vγ9Vδ2T cells, mainly focusing on the role of BTN3A as the pAg sensing molecule, as well as its potential impact on downstream events of the activation process.


Subject(s)
Antigens, CD/immunology , Butyrophilins/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , T-Lymphocytes/drug effects , Animals , Antigens, CD/drug effects , Butyrophilins/drug effects , Butyrophilins/immunology , Diphosphates/pharmacology , Humans , Phosphorylation/drug effects , T-Lymphocytes/immunology
18.
Eur J Immunol ; 47(12): 2137-2141, 2017 12.
Article in English | MEDLINE | ID: mdl-28741710

ABSTRACT

From several years, the anticancer effects of Vγ9 T lymphocytes make these cells good candidates for cancer immunotherapies. However, the proved efficacy of γδ Τ cell-based cancer immunotherapies in some clinical trials was minimized due to the inherent toxicity of IL-2, which is essential for the combination therapy with Phosphoantigen (PAg). Recently, we showed that IL-33, a γ chain receptor-independent cytokine, was able to induce the in vitro proliferation of PAg-activated Vγ9 T cells, which were fully functional expressing IFN-γ and TNF-α and showing in vitro anti-tumor cytotoxicity. We proposed IL-33 as an alternative to IL-2 for Vγ9 T cell-based cancer immunotherapies, and have therefore evaluated the efficacy of this cytokine in preclinical investigations. This study shows that human Vγ9 T cells are able to proliferate in a mouse model with the combination of PAg and rhIL-33, and that IL-33-expanded Vγ9 T cells can prevent tumor growth in a mouse lymphoma model.


Subject(s)
Immunotherapy/methods , Interleukin-33/pharmacology , Lymphoma/drug therapy , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays/methods , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Interleukin-33/genetics , Lymphoma/immunology , Lymphoma/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Recombinant Proteins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Burden/drug effects , Tumor Burden/immunology
19.
Cell Immunol ; 296(1): 31-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25991474

ABSTRACT

The γδ T cell lineage in humans remains much of an enigma due to the low number of defined antigens, the non-canonical ways in which these cells respond to their environment and difficulty in tracking this population in vivo. In this review, we survey a comparative evolutionary analysis of the primate V, D and J gene segments and contrast these findings with recent progress in defining antigen recognition by different populations of γδ T cells in humans. Signatures of both purifying and diversifying selection at the Vδ and Vγ gene loci are placed into context of Vδ1+ γδ T cell recognition of CD1d presenting different lipids, and Vγ 9Vδ2 T cell modulation by pyrophosphate-based phosphoantigens through the butyrophilins BTN3A. From this comparison, it is clear that co-evolution between γδ TCRs and these ligands is likely occurring, but the diversity inherent in these recombined receptors is an important feature in ligand surveillance.


Subject(s)
Evolution, Molecular , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD1d/immunology , Butyrophilins , Callithrix , Diphosphates/immunology , Humans , Ligands , Membrane Glycoproteins/immunology , Pan troglodytes , Receptors, Antigen, T-Cell, gamma-delta/genetics
20.
Front Immunol ; 5: 688, 2014.
Article in English | MEDLINE | ID: mdl-25657647

ABSTRACT

The predominant population of γδ T cells in human blood express a T cell receptor (TCR) composed of a Vγ9 (Vγ2 in an alternate nomenclature) and Vδ2 domains. These cells came into the limelight when it was discovered they can respond to certain microbial infections and tumorigenic cells through the detection of small, pyrophosphate containing organic molecules collectively called "phosphoantigens" or "pAgs." These molecules are intermediates in both eukaryotic and prokaryotic metabolic pathways. Chemical variants of these intermediates have been used in the clinic to treat a range of different cancers, however, directed optimization of these molecules requires a full understanding of their mechanism of action on target cells. We and others have identified a subclass of butyrophilin-related molecules (BTN3A1-3) that are directly involved in pAg sensing in the target cell, leading to engagement and activation of the T cell through the TCR. Our data and that of others support the pAg binding site to be the intracellular B30.2 domain of BTN3A1, which is the only isoform capable of mediating pAg-dependent stimulation of Vγ9Vδ2 T cells. Here, we review the data demonstrating pAg binding to the B30.2 domain and our studies of the structural conformations of the BTN3A extracellular domains. Finally, we synthesize a model linking binding of pAg to the intracellular domain with T cell detection via the extracellular domains in an "inside-out" signaling mechanism of the type characterized first for integrin molecule signaling. We also explore the role of Vγ9Vδ2 TCR variability in the CDR3 γ and δ loops and how this may modulate Vγ9Vδ2 cells as a population in surveillance of human health and disease.

SELECTION OF CITATIONS
SEARCH DETAIL