1
|
Lebrec H, Bui J, Clingan JM, Do J, Dubovsky J, Dragone L, Gibiansky E, Lam WY, Matsuda K, Mihalcik L, Ramsdell F, van der Vuurst de Vries AR, Xiao Y, Bluestone JA. Second generation CD2-targeting LFA-3 fusion protein SBT115301 to restore immune homeostasis in autoimmune disease. iScience 2025; 28:112447. [PMID: 40491961 PMCID: PMC12146614 DOI: 10.1016/j.isci.2025.112447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/14/2025] [Accepted: 04/11/2025] [Indexed: 06/11/2025] Open
Abstract
In autoimmunity, an imbalance of effector (Teff) and regulatory (Treg)T cells contributes to inflammation and tissue destruction. CD2, highly expressed on Teff and at lower levels on Treg and naive T cells (Tn), is an attractive target for depleting Teff at sites of inflammation. SBT115301 is a second generation CD2-targeting fusion protein containing the cognate receptor of CD2, lymphocyte function associated antigen-3 (LFA-3; CD58). In in vitro and in vivo studies, SBT115301 preferentially decreased CD2hi-expressing Teff cells compared to Treg and Tn. In a phase 1 clinical trial, SBT115301 selectively reduced memory T cells. SBT115301 was well tolerated aside from decreases of CD4+ T cells in some participants in the highest dose IM and IV cohorts. Anti-drug antibodies decreased exposure of SBT115301 in some participants without affecting the pharmacodynamics. These data support further study of SBT115301 as a monotherapy or in combination with other drugs in autoimmune indications.
Collapse
Affiliation(s)
- Herve Lebrec
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | - John Bui
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | - Jonathan M. Clingan
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | - Jason Do
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | - Jason Dubovsky
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | - Leonard Dragone
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | | | - Wing Yu Lam
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | - Katherine Matsuda
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | - Lauren Mihalcik
- Aclairo Pharmaceutical Development Group, Vienna, VA 22182, USA
| | - Frederick Ramsdell
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | | | - Yuanyuan Xiao
- Sonoma Biotherapeutics, South San Francisco, CA 94080, Seattle, WA 98119, USA
| | | |
Collapse
|
2
|
Lui Y, Ferreira Fernandes J, Vuong MT, Sharma S, Santos AM, Davis SJ. The Structural Biology of T-Cell Antigen Detection at Close Contacts. Immunol Rev 2025; 331:e70014. [PMID: 40181535 PMCID: PMC11969063 DOI: 10.1111/imr.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 04/05/2025]
Abstract
T cells physically interrogate their targets using tiny membrane protrusions called microvilli, forming junctions ~400 nm in diameter and ~ 15 nm deep, referred to as "close contacts". These contacts, which are stabilized by the binding of the small adhesion protein CD2 to its ligand, CD58 and locally exclude large proteins such as the phosphatase CD45, are the sites of antigen recognition by the T-cell receptor (TCR) and very early signaling by T cells. With our collaborators, we have characterized the molecular structures of several of the key proteins mediating these early events: i.e., CD2 and its ligands, CD45, the αβ- and γδ-TCRs, and the accessory proteins CD28, CTLA-4, and PD-1. Here, we review our structural work and the insights it offers into the early events underpinning T-cell responsiveness that take place in the confined space of the close contact. We reflect on the crucial roles that the structural organization and dimensions of these proteins are likely to have in determining the sequence of events leading to antigen recognition at close contacts and consider the general implications of the structural work for explanations of how immune receptor signaling is initiated.
Collapse
Affiliation(s)
- Yuan Lui
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - João Ferreira Fernandes
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Mai T. Vuong
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Sumana Sharma
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Ana Mafalda Santos
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Simon J. Davis
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| |
Collapse
|
3
|
Müller-Kirschbaum LC, Lodygin D, Odoardi F, Merlini A. Induction and Assessment of Activation in Rat Effector CD4 + Memory Th Cells. Methods Mol Biol 2025; 2904:145-158. [PMID: 40220232 DOI: 10.1007/978-1-0716-4414-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
T cell activation is a multi-step process that results in proliferation and cytokine secretion to direct protective and pathological immune responses. The activation process is highly coordinated and requires the integration of signals from antigen recognition via the T cell receptor (TCR), costimulatory molecules, and cytokines. This chapter outlines methods to induce T helper cell (Th cell) activation in vitro and assesses its functional outcomes, including cytokine transcription and production, expression of surface activation markers, and proliferation. Two primary activation protocols are described: antigen-specific activation by the cognate antigen and presenting cells, and antigen-non-specific stimulation via anti-CD3/CD28 antibodies. The former emulates physiological conditions, while the latter offers reproducibility. We will then describe protocols for quantifying transcriptional and protein-level responses upon T cell activation.
Collapse
Affiliation(s)
- Lukas C Müller-Kirschbaum
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Göttingen, Germany
| | - Dmitri Lodygin
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Göttingen, Germany
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Göttingen, Germany
| | - Arianna Merlini
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Göttingen, Germany.
| |
Collapse
|
4
|
Morales-Martínez M, Andón-García D, Patiño-Santiago KA, Parga-Ortega JM, Hernández-Hernández A, Aquino-Jarquin G, Patino-Lopez G. Identification of potential new T cell activation molecules: a Bioinformatic Approach. Sci Rep 2024; 14:22219. [PMID: 39333573 PMCID: PMC11436975 DOI: 10.1038/s41598-024-73003-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
T-cell activation is central for the initiation of T cell mediated adaptive immune response and is the result of the close communication between the Antigen Presenting Cell (APC) and the T lymphocyte. Although T-cell activation is currently well understood, and many intracellular pathways are well characterized, nevertheless new players are constantly identified, and this complements the known protein interactome. In this work we aimed to identify new proteins involved in T cell activation. We reviewed and analyzed results of microarray gene expression datasets reported in the public database GEO-NCBI. Using data from GSE136625, GSE50971, GSE13887, GSE11989 and GSE902 we performed different comparisons using R and other bioinformatic tools including GEO2R and we report here upregulated genes that have no previous reports in immune related functions and with potential participation upon T-cell activation. Our results indicate that RND3, SYT10, IgSF6 and PIN1 are potential new T-cell activation molecules.
Collapse
Affiliation(s)
- Mario Morales-Martínez
- Immunology and Proteomics Laboratory, Children's Hospital of Mexico, Mexico City, 06720, Mexico
| | - David Andón-García
- Immunology and Proteomics Laboratory, Children's Hospital of Mexico, Mexico City, 06720, Mexico
| | | | | | | | - Guillermo Aquino-Jarquin
- RNA Biology and Genome Editing Section, Genomics, Genetics, and Bioinformatics Research Laboratory, 'Federico Gómez' Children's Hospital of Mexico, Mexico City, 06720, Mexico
| | - Genaro Patino-Lopez
- Immunology and Proteomics Laboratory, Children's Hospital of Mexico, Mexico City, 06720, Mexico.
| |
Collapse
|
5
|
Nicolai CJ, Parker MH, Qin J, Tang W, Ulrich-Lewis JT, Gottschalk RJ, Cooper SE, Hernandez Lopez SA, Parrilla D, Mangio RS, Ericson NG, Brandes AH, Umuhoza S, Michels KR, McDonnell MM, Park LY, Shin S, Leung WH, Scharenberg AM, Kiem HP, Larson RP, Beitz LO, Ryu BY. In vivo CAR T-cell generation in nonhuman primates using lentiviral vectors displaying a multidomain fusion ligand. Blood 2024; 144:977-987. [PMID: 38861668 PMCID: PMC11406189 DOI: 10.1182/blood.2024024523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell therapies have demonstrated transformative efficacy in treating B-cell malignancies. However, high costs and manufacturing complexities hinder their widespread use. To overcome these hurdles, we have developed the VivoVec platform, a lentiviral vector capable of generating CAR T cells in vivo. Here, we describe the incorporation of T-cell activation and costimulatory signals onto the surface of VivoVec particles (VVPs) in the form of a multidomain fusion protein and show enhanced in vivo transduction and improved CAR T-cell antitumor functionality. Furthermore, in the absence of lymphodepleting chemotherapy, administration of VVPs into nonhuman primates resulted in the robust generation of anti-CD20 CAR T cells and the complete depletion of B cells for >10 weeks. These data validate the VivoVec platform in a translationally relevant model and support its transition into human clinical testing, offering a paradigm shift in the field of CAR T-cell therapies.
Collapse
Affiliation(s)
| | | | - Jim Qin
- Umoja Biopharma, Seattle, WA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Rogers J, Bajur AT, Salaita K, Spillane KM. Mechanical control of antigen detection and discrimination by T and B cell receptors. Biophys J 2024; 123:2234-2255. [PMID: 38794795 PMCID: PMC11331051 DOI: 10.1016/j.bpj.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
The adaptive immune response is orchestrated by just two cell types, T cells and B cells. Both cells possess the remarkable ability to recognize virtually any antigen through their respective antigen receptors-the T cell receptor (TCR) and B cell receptor (BCR). Despite extensive investigations into the biochemical signaling events triggered by antigen recognition in these cells, our ability to predict or control the outcome of T and B cell activation remains elusive. This challenge is compounded by the sensitivity of T and B cells to the biophysical properties of antigens and the cells presenting them-a phenomenon we are just beginning to understand. Recent insights underscore the central role of mechanical forces in this process, governing the conformation, signaling activity, and spatial organization of TCRs and BCRs within the cell membrane, ultimately eliciting distinct cellular responses. Traditionally, T cells and B cells have been studied independently, with researchers working in parallel to decipher the mechanisms of activation. While these investigations have unveiled many overlaps in how these cell types sense and respond to antigens, notable differences exist. To fully grasp their biology and harness it for therapeutic purposes, these distinctions must be considered. This review compares and contrasts the TCR and BCR, placing emphasis on the role of mechanical force in regulating the activity of both receptors to shape cellular and humoral adaptive immune responses.
Collapse
Affiliation(s)
- Jhordan Rogers
- Department of Chemistry, Emory University, Atlanta, Georgia
| | - Anna T Bajur
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.
| | - Katelyn M Spillane
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom; Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
7
|
Patel A, Andre V, Eguiguren SB, Barton MI, Burton J, Denham EM, Pettmann J, Mørch AM, Kutuzov MA, Siller-Farfán JA, Dustin ML, van der Merwe PA, Dushek O. Using CombiCells, a platform for titration and combinatorial display of cell surface ligands, to study T-cell antigen sensitivity modulation by accessory receptors. EMBO J 2024; 43:132-150. [PMID: 38177315 PMCID: PMC10897201 DOI: 10.1038/s44318-023-00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/05/2023] [Accepted: 11/10/2023] [Indexed: 01/06/2024] Open
Abstract
Understanding cellular decisions due to receptor-ligand interactions at cell-cell interfaces has been hampered by the difficulty of independently varying the surface density of multiple different ligands. Here, we express the synthetic binder protein SpyCatcher, designed to form spontaneous covalent bonds with interactors carrying a Spytag, on the cell surface. Using this, we show that addition of different concentrations and combinations of native Spytag-fused ligands allows for the combinatorial display of ligands on cells within minutes. We use this combinatorial display of cell surface ligands-called CombiCells-to assess T cell antigen sensitivity and the impact of T cell co-stimulation and co-inhibition receptors. We find that the T cell receptor (TCR) displayed greater sensitivity to peptides on major-histocompatibility complexes (pMHC) than synthetic chimeric antigen receptor (CARs) and bi-specific T cell engager (BiTEs) display to their target antigen, CD19. While TCR sensitivity was greatly enhanced by CD2/CD58 interactions, CAR sensitivity was primarily but more modestly enhanced by LFA-1/ICAM-1 interactions. Lastly, we show that PD-1/PD-L1 engagement inhibited T cell activation triggered solely by TCR/pMHC interactions, as well as the amplified activation induced by CD2 and CD28 co-stimulation. The ability to easily produce cells with different concentrations and combinations of ligands should accelerate the study of receptor-ligand interactions at cell-cell interfaces.
Collapse
Affiliation(s)
- Ashna Patel
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Violaine Andre
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | | | - Michael I Barton
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Jake Burton
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Eleanor M Denham
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
- EnaraBio Ltd, The Bellhouse Building, Oxford Science Park, Sanders Road, Oxford, OX44GD, UK
| | - Johannes Pettmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
- GlaxoSmithKline Pharmaceuticals, Rue de l'Institut 89, 1330, Rixensart, Belgium
| | - Alexander M Mørch
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Mikhail A Kutuzov
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | | | - Michael L Dustin
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | | | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| |
Collapse
|
8
|
Jenkins E, Körbel M, O'Brien-Ball C, McColl J, Chen KY, Kotowski M, Humphrey J, Lippert AH, Brouwer H, Santos AM, Lee SF, Davis SJ, Klenerman D. Antigen discrimination by T cells relies on size-constrained microvillar contact. Nat Commun 2023; 14:1611. [PMID: 36959206 PMCID: PMC10036606 DOI: 10.1038/s41467-023-36855-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 02/21/2023] [Indexed: 03/25/2023] Open
Abstract
T cells use finger-like protrusions called 'microvilli' to interrogate their targets, but why they do so is unknown. To form contacts, T cells must overcome the highly charged, barrier-like layer of large molecules forming a target cell's glycocalyx. Here, T cells are observed to use microvilli to breach a model glycocalyx barrier, forming numerous small (<0.5 μm diameter) contacts each of which is stabilized by the small adhesive protein CD2 expressed by the T cell, and excludes large proteins including CD45, allowing sensitive, antigen dependent TCR signaling. In the absence of the glycocalyx or when microvillar contact-size is increased by enhancing CD2 expression, strong signaling occurs that is no longer antigen dependent. Our observations suggest that, modulated by the opposing effects of the target cell glycocalyx and small adhesive proteins, the use of microvilli equips T cells with the ability to effect discriminatory receptor signaling.
Collapse
Affiliation(s)
- Edward Jenkins
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Markus Körbel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Caitlin O'Brien-Ball
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - James McColl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Kevin Y Chen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Mateusz Kotowski
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Jane Humphrey
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Anna H Lippert
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Heather Brouwer
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Steven F Lee
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
9
|
Burton J, Siller-Farfán JA, Pettmann J, Salzer B, Kutuzov M, van der Merwe PA, Dushek O. Inefficient exploitation of accessory receptors reduces the sensitivity of chimeric antigen receptors. Proc Natl Acad Sci U S A 2023; 120:e2216352120. [PMID: 36598945 PMCID: PMC9926289 DOI: 10.1073/pnas.2216352120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 10/24/2022] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptors (CARs) can redirect T cells to target abnormal cells, but their activity is limited by a profound defect in antigen sensitivity, the source of which remains unclear. Here, we show that CARs have a > 100-fold lower antigen sensitivity compared to the T cell receptor (TCR) when antigen is presented on antigen-presenting cells (APCs) but nearly identical sensitivity when antigen is presented as purified protein. We next systematically measured the impact of engaging important T cell accessory receptors (CD2, LFA-1, CD28, CD27, and 4-1BB) on antigen sensitivity by adding their purified ligands. Unexpectedly, we found that engaging CD2 or LFA-1 improved the antigen sensitivity of the TCR by 125- and 22-fold, respectively, but improved CAR sensitivity by only < 5-fold. This differential effect of CD2 and LFA-1 engagement on the TCR vs. CAR was confirmed using APCs. We found that sensitivity to antigen can be partially restored by fusing the CAR variable domains to the TCR CD3ε subunit (also known as a TRuC) and fully restored by exchanging the TCRαβ variable domains for those of the CAR (also known as STAR or HIT). Importantly, these improvements in TRuC and STAR/HIT sensitivity can be predicted by their enhanced ability to exploit CD2 and LFA-1. These findings demonstrate that the CAR sensitivity defect is a result of their inefficient exploitation of accessory receptors and suggest approaches to increase sensitivity.
Collapse
Affiliation(s)
- Jake Burton
- Sir William Dunn School of Pathology, University of Oxford, OX1 3REOxford, UK
| | | | - Johannes Pettmann
- Sir William Dunn School of Pathology, University of Oxford, OX1 3REOxford, UK
| | - Benjamin Salzer
- Sir William Dunn School of Pathology, University of Oxford, OX1 3REOxford, UK
| | - Mikhail Kutuzov
- Sir William Dunn School of Pathology, University of Oxford, OX1 3REOxford, UK
| | | | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, OX1 3REOxford, UK
| |
Collapse
|
10
|
Pandey PR, Rózycki B, Weikl TR. Molecular Dynamics Simulations of Immune Receptors and Ligands. Methods Mol Biol 2023; 2654:51-59. [PMID: 37106175 DOI: 10.1007/978-1-0716-3135-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Molecular dynamics simulations of immune receptor and ligand proteins in their native membrane environment allow to determine the orientational and structural variability of the proteins and protein complexes. The simulations complement the static, "membrane-free" structural information obtained from cryo-EM structures of transmembrane proteins in detergent micelles or from crystal structures of extracellular protein domains. Here we describe how to set up and perform simulations of transmembrane receptors, ligands, and receptor-ligand complexes.
Collapse
Affiliation(s)
- Prithvi R Pandey
- Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Bartosz Rózycki
- Institute of Physics of the Polish Academy of Sciences, Warszawa, Poland
| | - Thomas R Weikl
- Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.
| |
Collapse
|
11
|
Chann AS, Charnley M, Newton LM, Newbold A, Wiede F, Tiganis T, Humbert PO, Johnstone RW, Russell SM. Stepwise progression of β-selection during T cell development involves histone deacetylation. Life Sci Alliance 2022; 6:6/1/e202201645. [PMID: 36283704 PMCID: PMC9595210 DOI: 10.26508/lsa.202201645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 11/26/2022] Open
Abstract
During T cell development, the first step in creating a unique T cell receptor (TCR) is genetic recombination of the TCRβ chain. The quality of the new TCRβ is assessed at the β-selection checkpoint. Most cells fail this checkpoint and die, but the coordination of fate at the β-selection checkpoint is not yet understood. We shed new light on fate determination during β-selection using a selective inhibitor of histone deacetylase 6, ACY1215. ACY1215 disrupted the β-selection checkpoint. Characterising the basis for this disruption revealed a new, pivotal stage in β-selection, bookended by up-regulation of TCR co-receptors, CD28 and CD2, respectively. Within this "DN3bPre" stage, CD5 and Lef1 are up-regulated to reflect pre-TCR signalling, and their expression correlates with proliferation. These findings suggest a refined model of β-selection in which a coordinated increase in expression of pre-TCR, CD28, CD5 and Lef1 allows for modulating TCR signalling strength and culminates in the expression of CD2 to enable exit from the β-selection checkpoint.
Collapse
Affiliation(s)
- Anchi S Chann
- Optical Sciences Centre, School of Science, Swinburne University of Technology, Hawthorn, Australia,Peter MacCallum Cancer Centre, Melbourne, Australia,Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Mirren Charnley
- Optical Sciences Centre, School of Science, Swinburne University of Technology, Hawthorn, Australia,Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Lucas M Newton
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Andrea Newbold
- Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Florian Wiede
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Patrick O Humbert
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia,Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, Australia,Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Australia,Department of Clinical Pathology, University of Melbourne, Melbourne, Australia
| | - Ricky W Johnstone
- Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Sarah M Russell
- Optical Sciences Centre, School of Science, Swinburne University of Technology, Hawthorn, Australia .,Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
12
|
Li B, Lu Y, Zhong MC, Qian J, Li R, Davidson D, Tang Z, Zhu K, Argenty J, de Peredo AG, Malissen B, Roncagalli R, Veillette A. Cis interactions between CD2 and its ligands on T cells are required for T cell activation. Sci Immunol 2022; 7:eabn6373. [PMID: 35930657 DOI: 10.1126/sciimmunol.abn6373] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
CD2 is largely described to promote T cell activation when engaged by its ligands, CD48 in mice and CD58 in humans, that are present on antigen-presenting cells (APCs). However, both CD48 and CD58 are also expressed on T cells. By generating new knockout mouse strains lacking CD2 or CD48 in the C57BL/6 background, we determined that whereas CD2 was necessary on T cells for T cell activation, its ligand CD48 was not required on APCs. Rather, CD48 was also needed on T cells. One exception was during cytotoxicity, which required CD48 on T cells and APCs. Fluorescence resonance energy transfer (FRET) studies in nonimmune cells provided evidence that cis interactions between CD2 and CD48 existed within individual cells. CD2-CD48 interactions on T cells enabled more robust T cell receptor (TCR) signals, including protein tyrosine phosphorylation. Using T cells from a CD2 knock-in mouse in which a tag was inserted at the carboxyl terminus of CD2, mass spectrometry analyses revealed that the role of CD2 in T cell activation correlated with its ability to interact with components of the TCR complex and the protein tyrosine kinase Lck. CD2-CD58 provided a similar function in human T cells. Thus, our data imply that T cell-intrinsic cis interactions of CD2 with its ligands are required for TCR signaling and T cell activation. Interactions with ligands on APCs contribute during cytotoxicity.
Collapse
Affiliation(s)
- Bin Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Molecular Biology Program, University of Montréal, Montréal, Québec H3T 1J4, Canada
| | - Yan Lu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Ming-Chao Zhong
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Jin Qian
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Rui Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Dominique Davidson
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Zhenghai Tang
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Kaiwen Zhu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jérémy Argenty
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS UPS, Toulouse, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France.,Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Molecular Biology Program, University of Montréal, Montréal, Québec H3T 1J4, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| |
Collapse
|
13
|
Diethelm P, Schmitz I, Iten I, Kisielow J, Matsushita M, Kopf M. LCMV induced down-regulation of HVEM on anti-viral T cells is critical for an efficient effector response. Eur J Immunol 2022; 52:924-935. [PMID: 35344223 PMCID: PMC9321772 DOI: 10.1002/eji.202048569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/01/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
T‐cell responses against tumors and pathogens are critically shaped by cosignaling molecules providing a second signal. Interaction of herpes virus entry mediator (HVEM, CD270, TNFRSF14) with multiple ligands has been proposed to promote or inhibit T‐cell responses and inflammation, dependent on the context. In this study, we show that absence of HVEM did neither affect generation of effector nor maintenance of memory antiviral T cells and accordingly viral clearance upon acute and chronic lymphocytic choriomeningitis virus (LCMV) infection, due to potent HVEM downregulation during infection. Notably, overexpression of HVEM on virus‐specific CD8+ T cells resulted in a reduction of effector cells, whereas numbers of memory cells were increased. Overall, this study indicates that downregulation of HVEM driven by LCMV infection ensures an efficient acute response at the price of impaired formation of T‐cell memory.
Collapse
Affiliation(s)
- Patrizia Diethelm
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Iwana Schmitz
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Irina Iten
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Jan Kisielow
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Mai Matsushita
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| |
Collapse
|
14
|
Siokis A, Robert PA, Demetriou P, Kvalvaag A, Valvo S, Mayya V, Dustin ML, Meyer-Hermann M. Characterization of mechanisms positioning costimulatory complexes in immune synapses. iScience 2021; 24:103100. [PMID: 34622155 PMCID: PMC8479700 DOI: 10.1016/j.isci.2021.103100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/12/2021] [Accepted: 09/07/2021] [Indexed: 11/30/2022] Open
Abstract
Small immunoglobulin superfamily (sIGSF) adhesion complexes form a corolla of microdomains around an integrin ring and secretory core during immunological synapse (IS) formation. The corolla recruits and retains major costimulatory/checkpoint complexes, such as CD28, making forces that govern corolla formation of particular interest. Here, we investigated the mechanisms underlying molecular reorganization of CD2, an adhesion and costimulatory molecule of the sIGSF family during IS formation. Computer simulations showed passive distal exclusion of CD2 complexes under weak interactions with the ramified F-actin transport network. Attractive forces between CD2 and CD28 complexes relocate CD28 from the IS center to the corolla. Size-based sorting interactions with large glycocalyx components, such as CD45, or short-range CD2 self-attraction successfully explain the corolla 'petals.' This establishes a general simulation framework for complex pattern formation observed in cell-bilayer and cell-cell interfaces, and the suggestion of new therapeutic targets, where boosting or impairing characteristic pattern formation can be pivotal.
Collapse
Affiliation(s)
- Anastasios Siokis
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
| | - Philippe A. Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
| | - Philippos Demetriou
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Audun Kvalvaag
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Salvatore Valvo
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Viveka Mayya
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Michael L. Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig 38106, Germany
| |
Collapse
|
15
|
Liu S, Xu J, Wu J. The Role of Co-Signaling Molecules in Psoriasis and Their Implications for Targeted Treatment. Front Pharmacol 2021; 12:717042. [PMID: 34354596 PMCID: PMC8329336 DOI: 10.3389/fphar.2021.717042] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Psoriasis is a chronic, systemic immune-mediated inflammatory disease manifesting in the skin, joint or both. Co-signaling molecules are essential for determining the magnitude of the T cell response to the antigen. According to the function of co-signaling molecules, they can be divided into co-stimulatory molecules and co-inhibitory molecules. The role of co-signaling molecules in psoriasis is recognized, mainly including the co-stimulatory molecules CD28, CD40, OX40, CD27, DR3, LFA-1, and LFA-3 and the co-inhibitory molecules CTLA-4, PD-1, and TIM-3. They impact the pathological process of psoriasis by modulating the immune strength of T cells, regulating the production of cytokines or the differentiation of Tregs. In recent years, immunotherapies targeting co-signaling molecules have made significant progress and shown broad application prospects in psoriasis. This review aims to outline the possible role of co-signaling molecules in the pathogenesis of psoriasis and their potential application for the treatment of psoriasis.
Collapse
Affiliation(s)
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Pettmann J, Huhn A, Abu Shah E, Kutuzov MA, Wilson DB, Dustin ML, Davis SJ, van der Merwe PA, Dushek O. The discriminatory power of the T cell receptor. eLife 2021; 10:e67092. [PMID: 34030769 PMCID: PMC8219380 DOI: 10.7554/elife.67092] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/15/2021] [Indexed: 12/20/2022] Open
Abstract
T cells use their T cell receptors (TCRs) to discriminate between lower-affinity self and higher-affinity non-self peptides presented on major histocompatibility complex (pMHC) antigens. Although the discriminatory power of the TCR is widely believed to be near-perfect, technical difficulties have hampered efforts to precisely quantify it. Here, we describe a method for measuring very low TCR/pMHC affinities and use it to measure the discriminatory power of the TCR and the factors affecting it. We find that TCR discrimination, although enhanced compared with conventional cell-surface receptors, is imperfect: primary human T cells can respond to pMHC with affinities as low as KD ∼ 1 mM. The kinetic proofreading mechanism fit our data, providing the first estimates of both the time delay (2.8 s) and number of biochemical steps (2.67) that are consistent with the extraordinary sensitivity of antigen recognition. Our findings explain why self pMHC frequently induce autoimmune diseases and anti-tumour responses, and suggest ways to modify TCR discrimination.
Collapse
Affiliation(s)
- Johannes Pettmann
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
- Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Anna Huhn
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| | - Enas Abu Shah
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
- Kennedy Institute of Rheumatology, University of OxfordOxfordUnited Kingdom
| | - Mikhail A Kutuzov
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| | - Daniel B Wilson
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
- Boston University, Department of Mathematics and StatisticsBostonUnited States
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of OxfordOxfordUnited Kingdom
| | - Simon J Davis
- Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | | | - Omer Dushek
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
17
|
Różycki B, Weikl TR. Cooperative Stabilization of Close-Contact Zones Leads to Sensitivity and Selectivity in T-Cell Recognition. Cells 2021; 10:1023. [PMID: 33926103 PMCID: PMC8145674 DOI: 10.3390/cells10051023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/30/2022] Open
Abstract
T cells are sensitive to 1 to 10 foreign-peptide-MHC complexes among a vast majority of self-peptide-MHC complexes, and discriminate selectively between peptide-MHC complexes that differ not much in their binding affinity to T-cell receptors (TCRs). Quantitative models that aim to explain this sensitivity and selectivity largely focus on single TCR/peptide-MHC complexes, but T cell adhesion involves a multitude of different complexes. In this article, we demonstrate in a three-dimensional computational model of T-cell adhesion that the cooperative stabilization of close-contact zones is sensitive to one to three foreign-peptide-MHC complexes and occurs at a rather sharp threshold affinity of these complexes, which implies selectivity. In these close-contact zones with lateral extensions of hundred to several hundred nanometers, few TCR/foreign-peptide-MHC complexes and many TCR/self-peptide-MHC complexes are segregated from LFA-1/ICAM-1 complexes that form at larger membrane separations. Previous high-resolution microscopy experiments indicate that the sensitivity and selectivity in the formation of closed-contact zones reported here are relevant for T-cell recognition, because the stabilization of close-contact zones by foreign, agonist peptide-MHC complexes precedes T-cell signaling and activation in the experiments.
Collapse
Affiliation(s)
- Bartosz Różycki
- Institute of Physics, Polish Academy of Sciences, Al. Lotników 32/46, 02-668 Warsaw, Poland;
| | - Thomas R. Weikl
- Department of Theory and Bio-Systems, Max Planck Institut of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| |
Collapse
|
18
|
Rosetting T cells in Hodgkin lymphoma are activated by immunological synapse components HLA class II and CD58. Blood 2021; 136:2437-2441. [PMID: 32589698 DOI: 10.1182/blood.2020005546] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
A unique feature of Hodgkin lymphoma (HL) is the presence of CD4+ T cells that surround, protect, and promote survival of tumor cells. The adhesion molecules involved in this so-called T-cell rosetting are important components of the immunological synapse (IS). However, it is unknown whether this synapse is fully assembled and leads to T-cell activation by enabling interaction between the T-cell receptor (TCR) and human leukocyte antigen class II (HLA-II). We established a novel rosetting model by coculturing HLA-II-matched peripheral blood mononuclear cells with HL cell lines and showed IS formation with activation of rosetting T cells. HLA-II downregulation by class II transactivator knockout did not affect the extent of rosetting, but almost completely abrogated T-cell activation. Intriguingly, the level of CD58 expression correlated with the extent of rosette formation, and CD58 knockout or CD2 blockade reduced both rosette formation and T-cell activation. The extension of our findings to primary HL tissue by immunohistochemistry and proximity ligation assays showed interaction of CD2 with CD58 and of TCR-associated CD4 with HLA-II. In conclusion, T-cell rosetting in HL is established by formation of the IS, and activation of rosetting T cells critically depends on the interaction of both TCR-HLA-II and CD2-CD58.
Collapse
|
19
|
Castro-Sanchez P, Teagle AR, Prade S, Zamoyska R. Modulation of TCR Signaling by Tyrosine Phosphatases: From Autoimmunity to Immunotherapy. Front Cell Dev Biol 2020; 8:608747. [PMID: 33425916 PMCID: PMC7793860 DOI: 10.3389/fcell.2020.608747] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Early TCR signaling is dependent on rapid phosphorylation and dephosphorylation of multiple signaling and adaptor proteins, leading to T cell activation. This process is tightly regulated by an intricate web of interactions between kinases and phosphatases. A number of tyrosine phosphatases have been shown to modulate T cell responses and thus alter T cell fate by negatively regulating early TCR signaling. Mutations in some of these enzymes are associated with enhanced predisposition to autoimmunity in humans, and mouse models deficient in orthologous genes often show T cell hyper-activation. Therefore, phosphatases are emerging as potential targets in situations where it is desirable to enhance T cell responses, such as immune responses to tumors. In this review, we summarize the current knowledge about tyrosine phosphatases that regulate early TCR signaling and discuss their involvement in autoimmunity and their potential as targets for tumor immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Rose Zamoyska
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Kanatsu-Shinohara M, Chen G, Morimoto H, Shinohara T. CD2 is a surface marker for mouse and rat spermatogonial stem cells. J Reprod Dev 2020; 66:341-349. [PMID: 32213736 PMCID: PMC7470899 DOI: 10.1262/jrd.2020-019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The spermatogonial stem cell (SSC) population in testis is small, and the lack of SSC markers has severely handicapped research on these cells. During our attempt to identify
genes involved in SSC aging, we found that CD2 is expressed in cultured SSCs. Flow cytometric analysis and spermatogonial transplantation experiments showed that CD2 is expressed
in SSCs from mature adult mouse testes. Cultured SSCs transfected with short hairpin RNAs (shRNAs) against CD2 proliferated poorly and showed an increased frequency of apoptosis.
Moreover, functional analysis of transfected cells revealed impairment of SSC activity. Fluorescence activated cell sorting and spermatogonial transplantation experiments showed
that CD2 is expressed not only in mouse but also in rat SSCs. The results indicate that CD2 is a novel SSC surface marker conserved between mouse and rat SSCs.
Collapse
Affiliation(s)
- Mito Kanatsu-Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.,AMED-CREST, AMED, Tokyo 100-0004, Japan
| | - Guiying Chen
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Hiroko Morimoto
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.,AMED-CREST, AMED, Tokyo 100-0004, Japan
| |
Collapse
|
21
|
Abu-Shah E, Trendel N, Kruger P, Nguyen J, Pettmann J, Kutuzov M, Dushek O. Human CD8 + T Cells Exhibit a Shared Antigen Threshold for Different Effector Responses. THE JOURNAL OF IMMUNOLOGY 2020; 205:1503-1512. [PMID: 32817332 PMCID: PMC7477745 DOI: 10.4049/jimmunol.2000525] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022]
Abstract
CD8+ T cells produce TNF-α, IL-2, and IFN-γ with similar Ag thresholds. Costimulation decreases Ag thresholds similarly for different cytokines. A common rate-limiting switch downstream of the TCR can explain these findings.
T cells recognizing cognate pMHC Ags become activated to elicit a myriad of cellular responses, such as target cell killing and the secretion of different cytokines, that collectively contribute to adaptive immunity. These effector responses have been hypothesized to exhibit different Ag dose and affinity thresholds, suggesting that pathogen-specific information may be encoded within the nature of the Ag. In this study, using systematic experiments in a reductionist system, in which primary human CD8+ T cell blasts are stimulated by recombinant peptides presented on MHC Ag alone, we show that different inflammatory cytokines have comparable Ag dose thresholds across a 25,000-fold variation in affinity. Although costimulation by CD28, CD2, and CD27 increased cytokine production in this system, the Ag threshold remained comparable across different cytokines. When using primary human memory CD8+ T cells responding to autologous APCs, equivalent thresholds were also observed for different cytokines and killing. These findings imply a simple phenotypic model of TCR signaling in which multiple T cell responses share a common rate-limiting threshold and a conceptually simple model of CD8+ T cell Ag recognition, in which Ag dose and affinity do not provide any additional response-specific information.
Collapse
Affiliation(s)
- Enas Abu-Shah
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; and.,Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Nicola Trendel
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; and
| | - Philipp Kruger
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; and
| | - John Nguyen
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; and
| | - Johannes Pettmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; and
| | - Mikhail Kutuzov
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; and
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; and
| |
Collapse
|
22
|
Céspedes PF, Beckers D, Dustin ML, Sezgin E. Model membrane systems to reconstitute immune cell signaling. FEBS J 2020; 288:1070-1090. [DOI: 10.1111/febs.15488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/26/2020] [Accepted: 07/14/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Pablo F. Céspedes
- Kennedy Institute of Rheumatology Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences University of Oxford UK
| | - Daniel Beckers
- MRC Human Immunology Unit MRC Weatherall Institute of Molecular Medicine University of Oxford UK
| | - Michael L. Dustin
- Kennedy Institute of Rheumatology Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences University of Oxford UK
| | - Erdinc Sezgin
- MRC Human Immunology Unit MRC Weatherall Institute of Molecular Medicine University of Oxford UK
- Science for Life Laboratory Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| |
Collapse
|
23
|
Zurli V, Montecchi T, Heilig R, Poschke I, Volkmar M, Wimmer G, Boncompagni G, Turacchio G, D'Elios MM, Campoccia G, Resta N, Offringa R, Fischer R, Acuto O, Baldari CT, Kabanova A. Phosphoproteomics of CD2 signaling reveals AMPK-dependent regulation of lytic granule polarization in cytotoxic T cells. Sci Signal 2020; 13:13/631/eaaz1965. [PMID: 32398348 DOI: 10.1126/scisignal.aaz1965] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Understanding the costimulatory signaling that enhances the activity of cytotoxic T cells (CTLs) could identify potential targets for immunotherapy. Here, we report that CD2 costimulation plays a critical role in target cell killing by freshly isolated human CD8+ T cells, which represent a challenging but valuable model to gain insight into CTL biology. We found that CD2 stimulation critically enhanced signaling by the T cell receptor in the formation of functional immune synapses by promoting the polarization of lytic granules toward the microtubule-organizing center (MTOC). To gain insight into the underlying mechanism, we explored the CD2 signaling network by phosphoproteomics, which revealed 616 CD2-regulated phosphorylation events in 373 proteins implicated in the regulation of vesicular trafficking, cytoskeletal organization, autophagy, and metabolism. Signaling by the master metabolic regulator AMP-activated protein kinase (AMPK) was a critical node in the CD2 network, which promoted granule polarization toward the MTOC in CD8+ T cells. Granule trafficking was driven by active AMPK enriched on adjacent lysosomes, revealing previously uncharacterized signaling cross-talk between vesicular compartments in CD8+ T cells. Our results thus establish CD2 signaling as key for mediating cytotoxic killing and granule polarization in freshly isolated CD8+ T cells and strengthen the rationale to choose CD2 and AMPK as therapeutic targets to enhance CTL activity.
Collapse
Affiliation(s)
- Vanessa Zurli
- Department of Life Sciences, University of Siena, via Aldo Moro 2, Siena 53100, Italy
| | - Tommaso Montecchi
- Department of Life Sciences, University of Siena, via Aldo Moro 2, Siena 53100, Italy
| | - Raphael Heilig
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Isabel Poschke
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Michael Volkmar
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Giuliana Wimmer
- Department of Life Sciences, University of Siena, via Aldo Moro 2, Siena 53100, Italy
| | - Gioia Boncompagni
- Department of Life Sciences, University of Siena, via Aldo Moro 2, Siena 53100, Italy
| | - Gabriele Turacchio
- Institute of Biochemistry and Cell Biology, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Giuseppe Campoccia
- Department of Immune Hematology and Transfusion Medicine, University Hospital of Siena, viale Bracci 16, Siena 53100, Italy
| | - Nicoletta Resta
- Medical Genetics Unit, Department of Biomedical Sciences and Human Oncology, University of Bari, Policlinico Hospital, Piazza Giulio Cesare 11, Bari 70124, Italy
| | - Rienk Offringa
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany.,Department of Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 280, Heidelberg, 69120, Germany
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Oreste Acuto
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | | - Anna Kabanova
- Department of Life Sciences, University of Siena, via Aldo Moro 2, Siena 53100, Italy.
| |
Collapse
|
24
|
Figueiredo CR, Kalirai H, Sacco JJ, Azevedo RA, Duckworth A, Slupsky JR, Coulson JM, Coupland SE. Loss of BAP1 expression is associated with an immunosuppressive microenvironment in uveal melanoma, with implications for immunotherapy development. J Pathol 2020; 250:420-439. [PMID: 31960425 PMCID: PMC7216965 DOI: 10.1002/path.5384] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/28/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022]
Abstract
Immunotherapy using immune checkpoint inhibitors (ICIs) induces durable responses in many metastatic cancers. Metastatic uveal melanoma (mUM), typically occurring in the liver, is one of the most refractory tumours to ICIs and has dismal outcomes. Monosomy 3 (M3), polysomy 8q, and BAP1 loss in primary uveal melanoma (pUM) are associated with poor prognoses. The presence of tumour-infiltrating lymphocytes (TILs) within pUM and surrounding mUM - and some evidence of clinical responses to adoptive TIL transfer - strongly suggests that UMs are indeed immunogenic despite their low mutational burden. The mechanisms that suppress TILs in pUM and mUM are unknown. We show that BAP1 loss is correlated with upregulation of several genes associated with suppressive immune responses, some of which build an immune suppressive axis, including HLA-DR, CD38, and CD74. Further, single-cell analysis of pUM by mass cytometry confirmed the expression of these and other markers revealing important functions of infiltrating immune cells in UM, most being regulatory CD8+ T lymphocytes and tumour-associated macrophages (TAMs). Transcriptomic analysis of hepatic mUM revealed similar immune profiles to pUM with BAP1 loss, including the expression of IDO1. At the protein level, we observed TAMs and TILs entrapped within peritumoural fibrotic areas surrounding mUM, with increased expression of IDO1, PD-L1, and β-catenin (CTNNB1), suggesting tumour-driven immune exclusion and hence the immunotherapy resistance. These findings aid the understanding of how the immune response is organised in BAP1 - mUM, which will further enable functional validation of detected biomarkers and the development of focused immunotherapeutic approaches. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Carlos R Figueiredo
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Department of the Faculty of Medicine, MediCity Research Laboratory and Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Helen Kalirai
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Joseph J Sacco
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Department of Medical OncologyThe Clatterbridge Cancer CentreWirralUK
| | - Ricardo A Azevedo
- Department of Cancer BiologyThe University of Texas–MD Anderson Cancer CenterHoustonTXUSA
| | - Andrew Duckworth
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Joseph R Slupsky
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Judy M Coulson
- Department of Cellular and Molecular PhysiologyUniversity of LiverpoolLiverpoolUK
| | - Sarah E Coupland
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Liverpool Clinical LaboratoriesRoyal Liverpool University HospitalLiverpoolUK
| |
Collapse
|
25
|
Abstract
T cells initiate and regulate adaptive immune responses that can clear infections. To do this, they use their T cell receptors (TCRs) to continually scan the surfaces of other cells for cognate peptide antigens presented on major histocompatibility complexes (pMHCs). Experimental work has established that as few 1-10 pMHCs are sufficient to activate T cells. This sensitivity is remarkable in light of a number of factors, including the observation that the TCR and pMHC are short molecules relative to highly abundant long surface molecules, such as CD45, that can hinder initial binding, and moreover, the TCR/pMHC interaction is of weak affinity with solution lifetimes of approximately 1 second. Here, we review experimental and mathematical work that has contributed to uncovering molecular mechanisms of T cell sensitivity. We organize the mechanisms by where they act in the pathway to activate T cells, namely mechanisms that (a) promote TCR/pMHC binding, (b) induce rapid TCR signaling, and (c) amplify TCR signaling. We discuss work showing that high sensitivity reduces antigen specificity unless molecular feedbacks are invoked. We conclude by summarizing a number of open questions.
Collapse
Affiliation(s)
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Ellerman D. Bispecific T-cell engagers: Towards understanding variables influencing the in vitro potency and tumor selectivity and their modulation to enhance their efficacy and safety. Methods 2018; 154:102-117. [PMID: 30395966 DOI: 10.1016/j.ymeth.2018.10.026] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023] Open
Abstract
Bispecific molecules redirecting the cytotoxicity of T-cells are a growing class of therapeutics with numerous molecules being tested in clinical trials. However, it has been a long way since the proof of concept studies in the mid 1980's. In the process we have learnt about the impact of different variables related to the bispecific molecule and the target antigen on the potency of this type of drugs. This work reviews the insights gained and how that knowledge has been used to design more potent bispecific T-cell engagers. The more recent advancement of antibodies with this modality into safety studies in non-human primates and as well as in clinical studies has revealed potential toxicity liabilities for the mode of action. Modifications in existing antibody formats and new experimental molecules designed to mitigate these problems are discussed.
Collapse
|
27
|
Pettmann J, Santos AM, Dushek O, Davis SJ. Membrane Ultrastructure and T Cell Activation. Front Immunol 2018; 9:2152. [PMID: 30319617 PMCID: PMC6167458 DOI: 10.3389/fimmu.2018.02152] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/31/2018] [Indexed: 11/13/2022] Open
Abstract
The immune system serves as a crucial line of defense from infection and cancer, while also contributing to tissue homeostasis. Communication between immune cells is mediated by small soluble factors called cytokines, and also by direct cellular interactions. Cell-cell interactions are particularly important for T cell activation. T cells direct the adaptive immune response and therefore need to distinguish between self and foreign antigens. Even though decades have passed since the discovery of T cells, exactly why and how they are able to recognize and discriminate between antigens is still not fully understood. Early imaging of T cells was very successful in capturing the early stages of conjugate formation of T cells with antigen-presenting cells upon recognition of peptide-loaded major histocompatibility complexes by the T cell receptor (TCR). These studies lead to the discovery of a “supramolecular activation cluster” now known as the immunological synapse, followed by the identification of microclusters of TCRs formed upon receptor triggering, that eventually coalesce at the center of the synapse. New developments in light microscopy have since allowed attention to turn to the very earliest stages of T cell activation, and to resting cells, at high resolution. This includes single-molecule localization microscopy, which has been applied to the question of whether TCRs are pre-clustered on resting T cells, and lattice light-sheet microscopy that has enabled imaging of whole cells interacting with antigen-presenting cells. The utilization of lattice light-sheet microscopy has yielded important insights into structures called microvilli, which are small membrane protrusions on T cells that seem likely to have a large impact on T cell recognition and activation. Here we consider how imaging has shaped our thinking about T cell activation. We summarize recent findings obtained by applying more advanced microscopy techniques and discuss some of the limitations of these methods.
Collapse
Affiliation(s)
- Johannes Pettmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.,Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Simon J Davis
- Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
28
|
Muller J, Baeyens A, Dustin ML. Tumor Necrosis Factor Receptor Superfamily in T Cell Priming and Effector Function. Adv Immunol 2018; 140:21-57. [PMID: 30366518 DOI: 10.1016/bs.ai.2018.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tumor necrosis factor receptor superfamily (TNFRSF) and their ligands mediate lymphoid tissue development and homeostasis in addition to key aspects of innate and adaptive immune responses. T cells of the adaptive immune system express a number of TNFRSF members that are used to receive signals at different instructive stages and produce several tumor necrosis factor superfamily (TNFSF) members as effector molecules. There is also one example of a TNFRSF member serving as a ligand for negative regulatory checkpoint receptors. In most cases, the ligands in afferent and efferent phases are membrane proteins and thus the interaction with TNFRSF members must take place in immunological synapses and other modes of cell-cell interaction. A particular feature of the TNFRSF-mediated signaling is the prominent use of linear ubiquitin chains as scaffolds for signaling complexes that activate nuclear factor κ-B and Fos/Jun transcriptional regulators. This review will focus on the signaling mechanisms triggered by TNFRSF members in their role as costimulators of early and late phases of T cell instruction and the delivery mechanism of TNFSF members through the immunological synapses of helper and cytotoxic effector cells.
Collapse
Affiliation(s)
- James Muller
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States
| | - Audrey Baeyens
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States
| | - Michael L Dustin
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States; Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
29
|
Comrie WA, Burkhardt JK. Action and Traction: Cytoskeletal Control of Receptor Triggering at the Immunological Synapse. Front Immunol 2016; 7:68. [PMID: 27014258 PMCID: PMC4779853 DOI: 10.3389/fimmu.2016.00068] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 02/12/2016] [Indexed: 01/03/2023] Open
Abstract
It is well known that F-actin dynamics drive the micron-scale cell shape changes required for migration and immunological synapse (IS) formation. In addition, recent evidence points to a more intimate role for the actin cytoskeleton in promoting T cell activation. Mechanotransduction, the conversion of mechanical input into intracellular biochemical changes, is thought to play a critical role in several aspects of immunoreceptor triggering and downstream signal transduction. Multiple molecules associated with signaling events at the IS have been shown to respond to physical force, including the TCR, costimulatory molecules, adhesion molecules, and several downstream adapters. In at least some cases, it is clear that the relevant forces are exerted by dynamics of the T cell actomyosin cytoskeleton. Interestingly, there is evidence that the cytoskeleton of the antigen-presenting cell also plays an active role in T cell activation, by countering the molecular forces exerted by the T cell at the IS. Since actin polymerization is itself driven by TCR and costimulatory signaling pathways, a complex relationship exists between actin dynamics and receptor activation. This review will focus on recent advances in our understanding of the mechanosensitive aspects of T cell activation, paying specific attention to how F-actin-directed forces applied from both sides of the IS fit into current models of receptor triggering and activation.
Collapse
Affiliation(s)
- William A Comrie
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
30
|
Sun Y, Zhu X, Chen X, Liu H, Xu Y, Chu Y, Wang G, Liu X. The mediator subunit Med23 contributes to controlling T-cell activation and prevents autoimmunity. Nat Commun 2014; 5:5225. [PMID: 25301163 DOI: 10.1038/ncomms6225] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/10/2014] [Indexed: 12/11/2022] Open
Abstract
T-cell activation is critical for successful immune responses and is controlled at multiple levels. Although many changes of T-cell receptor-associated signalling molecules affect T-cell activation, the transcriptional mechanisms that control this process remain largely unknown. Here we find that T cell-specific deletion of the mediator subunit Med23 leads to hyperactivation of T cells and aged Med23-deficient mice exhibit an autoimmune syndrome. Med23 specifically and consistently promotes the transcription of multiple negative regulators of T-cell activation. In the absence of Med23, the T-cell activation threshold is lower, which results in enhanced antitumour T-cell function. Cumulatively, our data suggest that Med23 contributes to controlling T-cell activation at the transcriptional level and prevents the development of autoimmunity.
Collapse
Affiliation(s)
- Yang Sun
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoyan Zhu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xufeng Chen
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Haifeng Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Xu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yajing Chu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Gang Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaolong Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
31
|
Brady JL, Sutherland RM, Hancock M, Kitsoulis S, Lahoud MH, Phillips PM, Hawthorne WJ, d'Apice AJF, Cowan PJ, Harrison LC, O'Connell PJ, Lew AM. Anti-CD2 producing pig xenografts effect localized depletion of human T cells in a huSCID model. Xenotransplantation 2013; 20:100-9. [PMID: 23442186 DOI: 10.1111/xen.12025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/23/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND We investigated whether graft produced anti-human CD2, mediated by adenovirus (Adv) transduction of pig neonatal islet cell clusters (pNICC), would protect xenografts in a humanized mouse model from immune attack and whether such immunosuppression would remain local. METHODS A mouse anti-human CD2 Ab (CD2hb11) previously generated by us was genetically engineered to produce chimeric and humanized versions. The three forms of CD2hb11 were named dilimomab (mouse), diliximab (chimeric) and dilizumab (humanized). All 3 forms of CD2hb11 Ab were tested for their ability to bind CD3(+) human T cells and to inhibit a human anti-pig xenogeneic mixed lymphocyte reaction (MLR). They were administered systemically in a humanized mouse model in order to test their ability to deplete human CD3(+) T cells and whether they induced a cytokine storm. An adenoviral vector expressing diliximab was generated for transduction of pNICC. Humanized mice were transplanted with either control-transduced pNICC or diliximab-transduced pNICC and human T cells within grafts and spleens were enumerated by flow cytometry. RESULTS Dilimomab and diliximab inhibited a human anti-pig xenogeneic response but dilizumab did not. All 3 forms of CD2hb11 Ab bound human T cells in vitro though dilimomab and diliximab exhibited 300-fold higher avidity than dilizumab. All 3 anti-CD2 Abs could deplete human CD3(+) T cells in vivo in a humanized mouse model without inducing upregulation of activation markers or significant release of cytokines. Humanized mice transplanted with diliximab-transduced pNICC afforded depletion of CD3(+) T cells at the graft site leaving the peripheral immune system intact. CONCLUSIONS Local production of a single Ab against T cells can reduce graft infiltration at the xenograft site and may reduce the need for conventional, systemic immunosuppression.
Collapse
Affiliation(s)
- Jamie L Brady
- Walter & Eliza Hall Institute of Medical Research, Parkville, Vic. 3052, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The mechanisms that drive normal B cell differentiation and activation are frequently subverted by B cell lymphomas for their unlimited growth and survival. B cells are particularly prone to malignant transformation because the machinery used for antibody diversification can cause chromosomal translocations and oncogenic mutations. The advent of functional and structural genomics has greatly accelerated our understanding of oncogenic mechanisms in lymphomagenesis. The signaling pathways that normal B cells utilize to sense antigens are frequently derailed in B cell malignancies, leading to constitutive activation of prosurvival pathways. These malignancies co-opt transcriptional regulatory systems that characterize their normal B cell counterparts and frequently alter epigenetic regulators of chromatin structure and gene expression. These mechanistic insights are ushering in an era of targeted therapies for these cancers based on the principles of pathogenesis.
Collapse
Affiliation(s)
- Arthur L Shaffer
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
33
|
Abstract
CD2 is a cell adhesion molecule that mediates T-cell activation by binding to its ligand CD58 on antigen-presenting cells. Interaction between CD2 and CD58 or leukocyte function-associated antigen-3 (LFA-3) helps to optimize immune recognition facilitating contact between T lymphocytes and antigen-presenting cells. Modulation or inhibition of this interaction has been shown to be therapeutically useful in the treatment of autoimmune diseases. Antibodies and small molecules including peptides have been designed to modulate or disrupt the cell adhesion interactions due to CD2 and CD58. E-rosetting assay is a widely used method applied in the study of the modulation of CD2-CD58 interaction, which is either labor-intensive or radio-hazardous. In this chapter, we describe two methods that are used to study cell adhesion inhibition: (a) E-rosetting Assay and (b) Lymphocyte-epithelial assay. The second method, lymphocyte-epithelial assay, is a rapid and sensitive heterotypic cell adhesion assay for studying cell adhesion inhibition. The method relies on the CD2 expression on the surface of Jurkat cells and the CD58 expression on the surface of Caco-2 cells, which were confirmed by flow cytometry and ELISA studies respectively. This heterotypic cell adhesion assay described typically takes less than 4 h to perform, allows the evaluation of inhibitory activity of peptides/small molecules to modulate CD2-CD58 interaction in real cell system.
Collapse
|
34
|
Molecular identification of an MHC-independent ligand recognized by a human {alpha}/{beta} T-cell receptor. Blood 2011; 117:4816-25. [PMID: 21300979 DOI: 10.1182/blood-2010-11-317743] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
During an analysis of T-cell responses against human renal cell carcinoma (RCC), we identified a CD4(+) T-cell line that showed TCR-mediated recognition and lysis of nearly all RCC lines regardless of MHC type. We have now elucidated the nature of the ligand for this α/β TCR, and it contains no MHC-related moiety and does not involve classic peptide processing. First, matrix metalloproteinase 14 (MMP14) expressed on RCC cells releases membrane-bound TRAIL expressed by the T cell; then, soluble TRAIL binds to its receptor DR4 (TRAIL-R1), which is expressed on tumor cells, and this TRAIL-DR4 complex is recognized by the TCR through a complementarity-determining region 3α (CDR3α)-mediated interaction. Direct and specific antigen-TCR interaction was demonstrated when the immobilized recombinant TRAIL/DR4 complex stimulated the TCR. In addition, amino acid substitutions in the CDR3α of the TCR either obliterated or enhanced target-specific recognition. This description of the molecular nature of a non-MHC target structure recognized by a naturally occurring α/β TCR not only broadens our concept of what the TCR can recognize, but also raises the question of whether such a T cell could be of clinical utility against RCC.
Collapse
|
35
|
Abstract
T-cells are a vital type of white blood cell that circulate around our bodies, scanning for cellular abnormalities and infections. They recognise disease-associated antigens via a surface receptor called the T-cell antigen receptor (TCR). If there were a specific TCR for every single antigen, no mammal could possibly contain all the T-cells it needs. This is clearly absurd and suggests that T-cell recognition must, to the contrary, be highly degenerate. Yet highly promiscuous TCRs would appear to be equally impossible: they are bound to recognise self as well as non-self antigens. We review how contributions from mathematical analysis have helped to resolve the paradox of the promiscuous TCR. Combined experimental and theoretical work shows that TCR degeneracy is essentially dynamical in nature, and that the T-cell can differentially adjust its functional sensitivity to the salient epitope, "tuning up" sensitivity to the antigen associated with disease and "tuning down" sensitivity to antigens associated with healthy conditions. This paradigm of continual modulation affords the TCR repertoire, despite its limited numerical diversity, the flexibility to respond to almost any antigenic challenge while avoiding autoimmunity.
Collapse
Affiliation(s)
| | | | - Andrew K. Sewell
- Department of Medical Biochemistry and Immunology of the Cardiff University School of Medicine
| |
Collapse
|
36
|
Choudhuri K, Dustin ML. Signaling microdomains in T cells. FEBS Lett 2010; 584:4823-31. [PMID: 20965175 DOI: 10.1016/j.febslet.2010.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 10/12/2010] [Accepted: 10/13/2010] [Indexed: 01/22/2023]
Abstract
Sub-micron scale signaling domains induced in the plasma membrane of cells are thought to play important roles in signal transduction. In T cells, agonist MHC-peptide complexes induce small diffraction-limited domains enriched in T cell receptor (TCR) and signaling molecules. These microclusters serve as transient platforms for signal initiation and are required for sustained signaling in T cells, although each microcluster functions for only a couple of minutes. How they are formed, and what mechanisms promote and regulate signaling within TCR microclusters is largely unknown, although it is clear that TCR engagement and dynamic reorganization of cortical actin are involved. Here, we review current understanding of signaling within microclusters in T cells, and speculate on how these structures may form, initiate biochemical signals, and serve as sites of both signal integration and amplification, while also facilitating appropriate termination of TCR and related signaling.
Collapse
Affiliation(s)
- Kaushik Choudhuri
- Helen L and Martin S Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, New York, NY 10016, USA
| | | |
Collapse
|
37
|
Kim EO, Kim TJ, Kim N, Kim ST, Kumar V, Lee KM. Homotypic cell to cell cross-talk among human natural killer cells reveals differential and overlapping roles of 2B4 and CD2. J Biol Chem 2010; 285:41755-64. [PMID: 20813844 DOI: 10.1074/jbc.m110.137976] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human natural killer (NK) cells express an abundant level of 2B4 and CD2 on their surface. Their counter-receptors, CD48 and CD58, are also expressed on the NK cell surface, raising a question about the functional consequences of potential 2B4/CD48 and CD2/CD58 interactions. Using blocking antibodies specific to each receptor, we demonstrated that both 2B4/CD48 and CD2/CD58 interactions were essential for the development of NK effector functions: cytotoxicity and cytokine secretion. However, only 2B4/CD48, but not CD2/CD58, interactions were shown to be critical for the optimal NK cell proliferation in response to interleukin (IL)-2. IL-2-activated NK cells cultured in the absence of 2B4/CD48 or CD2/CD58 interactions were severely impaired for their ability to induce intracellular calcium mobilization and subsequent ERK activation upon tumor target exposure, suggesting that the early signaling pathway of NK receptors leading to impaired cytolysis and interferon (IFN)-γ secretion was inhibited. Nevertheless, these defects did not fully account for the reduced proliferation of NK cells in the absence of 2B4/CD48 interactions, because anti-CD2 or anti-CD58 monoclonal antibody (mAb)-treated NK cells, showing defective signaling and effector functions, displayed normal proliferation upon IL-2 stimulation. These results propose the signaling divergence between pathways leading to cell proliferation and cytotoxicity/cytokine release, which can be differentially regulated by 2B4 and CD2 during IL-2-driven NK cell activation. Collectively, these results reveal the importance of homotypic NK-to-NK cell cross-talk through 2B4/CD48 and CD2/CD58 pairs and further present their differential and overlapping roles in human NK cells.
Collapse
Affiliation(s)
- Eun-Ok Kim
- Department of Biochemistry, Division of Brain Korea 21 Program for Biomedical Science, Global Research Lab, Korea University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
38
|
Fooksman DR, Vardhana S, Vasiliver-Shamis G, Liese J, Blair DA, Waite J, Sacristán C, Victora GD, Zanin-Zhorov A, Dustin ML. Functional anatomy of T cell activation and synapse formation. Annu Rev Immunol 2010; 28:79-105. [PMID: 19968559 DOI: 10.1146/annurev-immunol-030409-101308] [Citation(s) in RCA: 361] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cell activation and function require a structured engagement of antigen-presenting cells. These cell contacts are characterized by two distinct dynamics in vivo: transient contacts resulting from promigratory junctions called immunological kinapses or prolonged contacts from stable junctions called immunological synapses. Kinapses operate in the steady state to allow referencing to self-peptide-MHC (pMHC) and searching for pathogen-derived pMHC. Synapses are induced by T cell receptor (TCR) interactions with agonist pMHC under specific conditions and correlate with robust immune responses that generate effector and memory T cells. High-resolution imaging has revealed that the synapse is highly coordinated, integrating cell adhesion, TCR recognition of pMHC complexes, and an array of activating and inhibitory ligands to promote or prevent T cell signaling. In this review, we examine the molecular components, geometry, and timing underlying kinapses and synapses. We integrate recent molecular and physiological data to provide a synthesis and suggest ways forward.
Collapse
Affiliation(s)
- David R Fooksman
- Department of Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, 10016, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kaizuka Y, Douglass AD, Vardhana S, Dustin ML, Vale RD. The coreceptor CD2 uses plasma membrane microdomains to transduce signals in T cells. J Cell Biol 2009; 185:521-34. [PMID: 19398758 PMCID: PMC2700390 DOI: 10.1083/jcb.200809136] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Accepted: 04/08/2009] [Indexed: 01/09/2023] Open
Abstract
The interaction between a T cell and an antigen-presenting cell (APC) can trigger a signaling response that leads to T cell activation. Prior studies have shown that ligation of the T cell receptor (TCR) triggers a signaling cascade that proceeds through the coalescence of TCR and various signaling molecules (e.g., the kinase Lck and adaptor protein LAT [linker for T cell activation]) into microdomains on the plasma membrane. In this study, we investigated another ligand-receptor interaction (CD58-CD2) that facilities T cell activation using a model system consisting of Jurkat T cells interacting with a planar lipid bilayer that mimics an APC. We show that the binding of CD58 to CD2, in the absence of TCR activation, also induces signaling through the actin-dependent coalescence of signaling molecules (including TCR-zeta chain, Lck, and LAT) into microdomains. When simultaneously activated, TCR and CD2 initially colocalize in small microdomains but then partition into separate zones; this spatial segregation may enable the two receptors to enhance signaling synergistically. Our results show that two structurally distinct receptors both induce a rapid spatial reorganization of molecules in the plasma membrane, suggesting a model for how local increases in the concentration of signaling molecules can trigger T cell signaling.
Collapse
Affiliation(s)
- Yoshihisa Kaizuka
- The Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143
| | - Adam D. Douglass
- The Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143
| | - Santosh Vardhana
- Program in Molecular Pathogenesis, Helen and Martin Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Michael L. Dustin
- Program in Molecular Pathogenesis, Helen and Martin Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Ronald D. Vale
- The Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
40
|
Giddu S, Subramanian V, Yoon HS, Satyanarayanajois SD. Design of beta-hairpin peptides for modulation of cell adhesion by beta-turn constraint. J Med Chem 2009; 52:726-36. [PMID: 19123855 DOI: 10.1021/jm8008212] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The CD2-CD58 interaction in immune regulation and disease pathology has provided new targets for developing potential immunosuppressive agents. In the present study, we report the introduction of constraints to generate beta-hairpin structures from the strand sequences of CD2 protein. The beta-hairpin structures were induced in the designed peptides by introducing Pro-Gly sequences in the peptides. Results from NMR and MD simulation indicated that the peptides exhibited beta-turn structure at the X-Pro-Gly-Y sequence and formed the beta-hairpin structure in solution. The ability of these peptides to inhibit cell adhesion was evaluated by two cell adhesion assays. Among the peptides studied (1-4) (P1-P4), peptides 2-4 were able to inhibit cell adhesion between Jurkat cells and SRBC nearly 50% at 180 microM, and 80% inhibition between Jurkat cells and Caco-2 cells was seen at 90 microM. Peptide 1 did not show significant inhibition activity compared to control.
Collapse
Affiliation(s)
- Sumana Giddu
- Department of Basic Pharmaceutical Sciences, University of Louisiana at Monroe, 700 University Avenue, Monroe, Louisiana 71209, USA
| | | | | | | |
Collapse
|
41
|
Chlewicki LK, Velikovsky CA, Balakrishnan V, Mariuzza RA, Kumar V. Molecular basis of the dual functions of 2B4 (CD244). THE JOURNAL OF IMMUNOLOGY 2008; 180:8159-67. [PMID: 18523281 DOI: 10.4049/jimmunol.180.12.8159] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
2B4 belongs to the CD2 family of molecules and is expressed on all NK, gammadelta, and memory CD8(+) (alphabeta) T cells. The murine NK receptor 2B4 exhibits both inhibitory and activating functions, whereas human 2B4 has been reported to be an activating molecule. How murine 2B4 can act both as an activating and inhibitory receptor and what distinguishes its function from human 2B4 have remained largely unknown. We use here a model system that allows the study of human and murine 2B4 under identical and controlled conditions. These studies reveal that both human and mouse 2B4 can activate or inhibit NK cells. We show here that the level of 2B4 expression and the degree of 2B4 cross-linking play a significant role in the regulation of signaling lymphocyte activation molecule-associated protein-mediated activation by 2B4. A high level of 2B4 expression, heavy cross-linking, and relative paucity of signaling lymphocyte activation molecule-associated protein promote inhibitory function. Our studies demonstrate how a single receptor can have opposing function depending on the degree of receptor expression, extent of its ligation, and the relative abundance of certain adaptor molecules. Because the levels of 2B4 and CD48 are dynamically regulated, these findings have implications for the regulation of NK cell function.
Collapse
|
42
|
Pawlowski NN, Struck D, Grollich K, Kuhl AA, Zeitz M, Liesenfeld O, Hoffmann JC. CD2 deficiency partially prevents small bowel inflammation and improves parasite control in murine Toxoplasma gondii infection. World J Gastroenterol 2007; 13:4207-13. [PMID: 17696249 PMCID: PMC4250619 DOI: 10.3748/wjg.v13.i31.4207] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether bowel inflammation and/or parasite control is altered in the absence of the T cell adhesion molecule CD2.
METHODS: Wildtype (WT) and CD2 deficient (CD2-/-) mice were infected with 100 cysts of Toxoplasma gondii (T. gondii) (ME49) by gavage. On d 7 after infection mice were killed. Necrosis and the number of parasites/cm ileum were determined. Cytokine levels of stimulated cells as well as sera were evaluated. Secondly, survival of WT vs CD2-/- mice was analysed using Kaplan-Meier analysis.
RESULTS: CD2-/- mice survived longer than WT mice (mean: 23.5 vs 7.1 d, P = 0.001). Further, CD2-/- mice showed less weight loss and less ileal inflammation than WT mice at d 7 post infection. In addition, the number of parasites in the ileum was significantly lower in CD2-/- mice than in WT mice (88 ± 12 vs 349 ± 58 cm, P < 0.01). This was paralleled by lower production of IFN-γ and IL-6 from TLA-stimulated mLN cells and increased IFN-γ production by splenocytes.
CONCLUSION: CD2 deficient mice are more resistant to T. gondii infection than WT mice. In contrast to most current immunosuppressive or biological therapies CD2 deficiency reduces intestinal inflammation and at the same time helps to control infection.
Collapse
Affiliation(s)
- Nina-N Pawlowski
- Medizinische Klinik I, St. Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Liu J, Li C, Ke S, Satyanarayanajois SD. Structure-based rational design of beta-hairpin peptides from discontinuous epitopes of cluster of differentiation 2 (CD2) protein to modulate cell adhesion interaction. J Med Chem 2007; 50:4038-47. [PMID: 17658775 DOI: 10.1021/jm0700868] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Modulation or inhibition of interaction of cluster of differentiation (CD) adhesion molecules CD2-CD58 has been shown to be therapeutically useful. The analysis of the crystal structure of CD2 complexed with CD58 was carried out to define the epitopes that are important for the interaction of the two proteins. The crystal structure of CD2 indicated that the interaction surface of CD2 with CD58 has two beta-strand structures (F and C strands) with charged residues. On the basis of the crystal structure of the complex CD2-CD58, we have designed beta-hairpin peptides from the beta-strand region of CD2 by conjugating the discontinuous sequences in the protein. The peptides were modeled by molecular dynamics simulation, and their inhibitory activities were evaluated in vitro using two heterotypic cell adhesion assays, E-rosetting and lymphocyte-epithelial cell adhesion assays. Results indicated that 12- and 14-residue conjugate cyclic peptides cKS12 and cDD14 exhibited 60% and 50% inhibition activity, respectively, at 90 microM.
Collapse
Affiliation(s)
- Jining Liu
- Department of Pharmacy, National University of Singapore, 117543, Singapore
| | | | | | | |
Collapse
|
44
|
Chamian F, Lin SL, Lee E, Kikuchi T, Gilleaudeau P, Sullivan-Whalen M, Cardinale I, Khatcherian A, Novitskaya I, Wittkowski KM, Krueger JG, Lowes MA. Alefacept (anti-CD2) causes a selective reduction in circulating effector memory T cells (Tem) and relative preservation of central memory T cells (Tcm) in psoriasis. J Transl Med 2007; 5:27. [PMID: 17555598 PMCID: PMC1906741 DOI: 10.1186/1479-5876-5-27] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 06/07/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alefacept (anti-CD2) biological therapy selectively targets effector memory T cells (Tem) in psoriasis vulgaris, a model Type 1 autoimmune disease. METHODS Circulating leukocytes were phenotyped in patients receiving alefacept for moderate to severe psoriasis. RESULTS In all patients, this treatment caused a preferential decrease in effector memory T cells (CCR7- CD45RA-) (mean 63% reduction) for both CD4+ and CD8+ Tem, while central memory T cells (Tcm) (CCR7+CD45RA-) were less affected, and naïve T cells (CCR7+CD45RA+) were relatively spared. Circulating CD8+ effector T cells and Type 1 T cells (IFN-gamma-producing) were also significantly reduced. CONCLUSION Alefacept causes a selective reduction in circulating effector memory T cells (Tem) and relative preservation of central memory T cells (Tcm) in psoriasis.
Collapse
Affiliation(s)
- Francesca Chamian
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Shao-Lee Lin
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Edmund Lee
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Toyoko Kikuchi
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Patricia Gilleaudeau
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Mary Sullivan-Whalen
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Irma Cardinale
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Artemis Khatcherian
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Inna Novitskaya
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Knut M Wittkowski
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Michelle A Lowes
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| |
Collapse
|
45
|
Haider AS, Lowes MA, Gardner H, Bandaru R, Darabi K, Chamian F, Kikuchi T, Gilleaudeau P, Whalen MS, Cardinale I, Novitskaya I, Krueger JG. Novel Insight into the Agonistic Mechanism of Alefacept In Vivo: Differentially Expressed Genes May Serve as Biomarkers of Response in Psoriasis Patients. THE JOURNAL OF IMMUNOLOGY 2007; 178:7442-9. [PMID: 17513795 DOI: 10.4049/jimmunol.178.11.7442] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Alefacept is an LFA3-Ig fusion protein that binds to CD2 and is thought to inhibit T cell activation by antagonism of CD2 signaling or by lysis of CD2(+) cells. Alefacept is potential future therapeutic for organ transplant recipients or graft-vs-host disease and is an approved therapeutic for psoriasis vulgaris, which is a T cell-mediated inflammatory disease. However, alefacept improves psoriasis in only approximately 50% of patients treated for 12 wk. We studied the immunologic effects of alefacept in a group of psoriasis patients during treatment. We found that T cells, especially CD8(+) T cells, were rapidly decreased in the peripheral circulation. Decreases in circulating T cells were not associated with induced apoptosis. Unexpectedly, in addition to suppression of inflammatory genes, we found a marked induction of mRNAs for STAT1, IL-8, and monokine induced by IFN-gamma during the first day of treatment in PBMC. We confirmed the agonistic effects of alefacept in PBMC in vitro, which were similar to CD3/CD28 ligation on T cells. These data establish that alefacept activates gene expression programs in leukocytes and suggest that its therapeutic action may be as a mixed agonist/antagonist. Furthermore, responding patients to alefacept treatment show unique patterns of gene modulation. Whereas alefacept down-regulated TCRs CD3D and CD2 in responders, nonresponders reveal a higher expression of T cell activation genes such as CD69 in pretreatment PBMC. These finding suggest a potential basis for categorizing responders vs nonresponders at an early time point in treatment or before treatment of a broad range of proinflammatory diseases. This study 1) establishes alefacept as a novel CD2 agonist molecule for induction of leukocyte activation genes (prior work proposed its mechanism as a CD2 antagonist) and 2) that differential activation of genes may categorize clinical responders to this agent, critical for cost-effective use of this drug.
Collapse
Affiliation(s)
- Asifa S Haider
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kearney A, Avramovic A, Castro MAA, Carmo AM, Davis SJ, van der Merwe PA. The contribution of conformational adjustments and long-range electrostatic forces to the CD2/CD58 interaction. J Biol Chem 2007; 282:13160-6. [PMID: 17344209 PMCID: PMC2771598 DOI: 10.1074/jbc.m700829200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CD2 is a T cell surface molecule that enhances T and natural killer cell function by binding its ligands CD58 (humans) and CD48 (rodents) on antigen-presenting or target cells. Here we show that the CD2/CD58 interaction is enthalpically driven and accompanied by unfavorable entropic changes. Taken together with structural studies, this indicates that binding is accompanied by energetically significant conformational adjustments. Despite having a highly charged binding interface, neither the affinity nor the rate constants of the CD2/CD58 interaction were affected by changes in ionic strength, indicating that long-range electrostatic forces make no net contribution to binding.
Collapse
Affiliation(s)
- Alice Kearney
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | | | | | | | | | | |
Collapse
|
47
|
Kittner JM, Jacobs R, Buyny S, Peest D, Stoll M, Schmidt RE. Adult onset of T-cell deficiency with impaired CD2 expression complicated by Rhodococcus infection: a case report. Ann Allergy Asthma Immunol 2007; 98:294-8. [PMID: 17378264 DOI: 10.1016/s1081-1206(10)60722-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND The functional importance of CD2 in vivo is currently the subject of discussion. OBJECTIVE To describe a 47-year-old white man with systemic Rhodococcus infection, a rarely observed opportunistic disease, secondary to severe lymphopenia. METHODS We extensively characterized lymphocyte phenotype and function. RESULTS Both CD4+ and CD8+ T cells were severely diminished, with a particular reduction in alpha:beta T cells. Human immunodeficiency virus infection was excluded. CD2 expression was decreased not only on T cells but also on nonaffected natural killer cells. Production of interferon-gamma interleukin 2, and tumor necrosis factor a was normal. Neither B-cell numbers nor humoral immune responses were affected. In addition, adhesion molecules CD11a, CD54, and CD154 were normally expressed, as were the costimulatory molecules CD28, CD80, and CD86. CONCLUSIONS We hypothesize that prolonged disturbance of CD2 expression led to an acquired severe cellular immunodeficiency. This underlines the importance of CD2 in vivo, where it may play a role in the fine regulation of T-cell proliferation.
Collapse
Affiliation(s)
- Jens Martin Kittner
- Department of Clinical Immunology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | |
Collapse
|
48
|
Zhu DM, Dustin ML, Cairo CW, Thatte HS, Golan DE. Mechanisms of Cellular Avidity Regulation in CD2-CD58-Mediated T Cell Adhesion. ACS Chem Biol 2006; 1:649-58. [PMID: 17168569 DOI: 10.1021/cb6002515] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The CD2 receptor on T lymphocytes is essential for T cell adhesion and stimulation by antigen presenting cells (APCs). Blockade of CD2 function is immunosuppressive in both model systems and humans, indicating the importance of CD2 for the cellular immune response. Although the affinity of the molecular interaction between CD2 and its counter-receptor, CD58, is relatively low when measured in solution, this interaction mediates tight adhesion within the 2D cell-cell interface. To understand the mechanisms responsible for regulating the avidity of the CD2-CD58 interaction, we measured the number, affinity, and lateral mobility of CD2 molecules on resting and activated T cells. Cell activation caused a 1.5-fold increase in the number of CD2 sites on the cell surface, and the 2D affinity of CD2 for CD58 increased by 2.5-fold. The combination of T cell activation and CD2 ligation to CD58 decreased the laterally mobile fraction of the ligated CD2. Together, these changes would substantially enhance CD2 avidity and strengthen T cell-APC adhesion. The change in CD2 mobile fraction suggests that the cell uses cytoskeletal regulators to immobilize the receptor selectively at the site of contact with surfaces expressing CD58. Our observations are consistent with a model in which T cell activation initially induces increased CD2 2D affinity, cell surface receptor expression, and lateral mobility, allowing the CD2 molecules to diffuse to sites of contact with CD58-bearing APCs. Subsequently, T cell activation causes the CD58-bound CD2 to be recognized and immobilized at sites of cell-cell contact, thereby strengthening T cell-APC adhesion.
Collapse
Affiliation(s)
- De-Min Zhu
- Departments of Biological Chemistry and Molecular Pharmacology, Surgery, and Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
49
|
Bendle GM, Holler A, Downs AM, Xue SA, Stauss HJ. Broadly expressed tumour-associated proteins as targets for cytotoxic T lymphocyte-based cancer immunotherapy. Expert Opin Biol Ther 2006; 5:1183-92. [PMID: 16120049 DOI: 10.1517/14712598.5.9.1183] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
T cell-based antigen-specific immunotherapy targeting self-proteins aberrantly expressed in many tumours offers the potential for widely applicable cancer immunotherapy, but carries the risk of autoimmunity. Immunological tolerance represents an inherent limitation of cancer vaccines targeting such broadly expressed tumour-associated proteins. Therefore, strategies to circumvent T cell tolerance have been developed and, when combined with T cell receptor (TCR) gene transfer technology, can generate highly avid tumour-reactive patient cytotoxic T lymphocytes (CTLs) specific for peptide epitopes of tumour-associated proteins. This review analyses the level of tolerance to broadly expressed tumour-associated proteins in the autologous T cell repertoire, assesses strategies that have been developed to circumvent T cell tolerance to such antigens, and evaluates the prospects for effective immunotherapy targeting broadly expressed tumour-associated proteins.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Clinical Trials as Topic
- Genetic Therapy
- Humans
- Immune Tolerance
- Immunotherapy/methods
- Models, Animal
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Proto-Oncogene Proteins c-mdm2/immunology
- Proto-Oncogene Proteins c-mdm2/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/transplantation
Collapse
Affiliation(s)
- Gavin M Bendle
- Royal Free Hospital, Department of Immunology and Molecular Pathology, University College London, London, NW3 2QG, UK
| | | | | | | | | |
Collapse
|
50
|
Evans EJ, Castro MAA, O'Brien R, Kearney A, Walsh H, Sparks LM, Tucknott MG, Davies EA, Carmo AM, van der Merwe PA, Stuart DI, Jones EY, Ladbury JE, Ikemizu S, Davis SJ. Crystal structure and binding properties of the CD2 and CD244 (2B4)-binding protein, CD48. J Biol Chem 2006; 281:29309-20. [PMID: 16803907 DOI: 10.1074/jbc.m601314200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The structural analysis of surface proteins belonging to the CD2 subset of the immunoglobulin superfamily has yielded important insights into transient cellular interactions. In mice and rats, CD2 and CD244 (2B4), which are expressed predominantly on T cells and natural killer cells, respectively, bind the same, broadly expressed ligand, CD48. Structures of CD2 and CD244 have been solved previously, and we now present the structure of the receptor-binding domain of rat CD48. The receptor-binding surface of CD48 is unusually flat, as in the case of rat CD2, and shares a high degree of electrostatic complementarity with the equivalent surface of CD2. The relatively simple arrangement of charged residues and this flat topology explain why CD48 cross-reacts with CD2 and CD244 and, in rats, with the CD244-related protein, 2B4R. Comparisons of modeled complexes of CD2 and CD48 with the complex of human CD2 and CD58 are suggestive of there being substantial plasticity in the topology of ligand binding by CD2. Thermodynamic analysis of the native CD48-CD2 interaction indicates that binding is driven by equivalent, weak enthalpic and entropic effects, in contrast to the human CD2-CD58 interaction, for which there is a large entropic barrier. Overall, the structural and biophysical comparisons of the CD2 homologues suggest that the evolutionary diversification of interacting cell surface proteins is rapid and constrained only by the requirement that binding remains weak and specific.
Collapse
Affiliation(s)
- Edward J Evans
- Nuffield Department of Clinical Medicine, The University of Oxford and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|