1
|
Minguet S, Maus MV, Schamel WW. From TCR fundamental research to innovative chimeric antigen receptor design. Nat Rev Immunol 2024:10.1038/s41577-024-01093-7. [PMID: 39433885 DOI: 10.1038/s41577-024-01093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/23/2024]
Abstract
Engineered T cells that express chimeric antigen receptors (CARs) have transformed the treatment of haematological cancers. CARs combine the tumour-antigen-binding function of antibodies with the signalling functions of the T cell receptor (TCR) ζ chain and co-stimulatory receptors. The resulting constructs aim to mimic the TCR-based and co-receptor-based activation of T cells. Although these have been successful for some types of cancer, new CAR formats are needed, to limit side effects and broaden their use to solid cancers. Insights into the mechanisms of TCR signalling, including the identification of signalling motifs that are not present in the TCR ζ chain and mechanistic insights in TCR activation, have enabled the development of CAR formats that outcompete the current CARs in preclinical mouse models and clinical trials. In this Perspective, we explore the mechanistic rationale behind new CAR designs.
Collapse
Affiliation(s)
- Susana Minguet
- Signalling Research Centers BIOSS and CIBSS, Freiburg, Germany.
- Department of Synthetic Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Marcela V Maus
- Cellular Immunotherapy Program and Krantz Family Center for Cancer Research, Mass General Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wolfgang W Schamel
- Signalling Research Centers BIOSS and CIBSS, Freiburg, Germany.
- Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
2
|
Sim MJW, Hanada KI, Stotz Z, Yu Z, Lu J, Brennan P, Quastel M, Gillespie GM, Long EO, Yang JC, Sun PD. Identification and structural characterization of a mutant KRAS-G12V specific TCR restricted by HLA-A3. Eur J Immunol 2024; 54:e2451079. [PMID: 39030753 DOI: 10.1002/eji.202451079] [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: 02/14/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/22/2024]
Abstract
Mutations in KRAS are some of the most common across multiple cancer types and are thus attractive targets for therapy. Recent studies demonstrated that mutant KRAS generates immunogenic neoantigens that are targetable by adoptive T-cell therapy in metastatic diseases. To expand mutant KRAS-specific immunotherapies, it is critical to identify additional HLA-I allotypes that can present KRAS neoantigens and their cognate T-cell receptors (TCR). Here, we identified a murine TCR specific to a KRAS-G12V neoantigen (7VVVGAVGVGK16) using a vaccination approach with transgenic mice expressing HLA-A*03:01 (HLA-A3). This TCR demonstrated exquisite specificity for mutant G12V and not WT KRAS peptides. To investigate the molecular basis for neoantigen recognition by this TCR, we determined its structure in complex with HLA-A3(G12V). G12V-TCR CDR3β and CDR1β formed a hydrophobic pocket to interact with p6 Val of the G12V but not the WT KRAS peptide. To improve the tumor sensitivity of this TCR, we designed rational substitutions to improve TCR:HLA-A3 contacts. Two substitutions exhibited modest improvements in TCR binding avidity to HLA-A3 (G12V) but did not sufficiently improve T-cell sensitivity for further clinical development. Our study provides mechanistic insight into how TCRs detect neoantigens and reveals the challenges in targeting KRAS-G12V mutations.
Collapse
Affiliation(s)
- Malcolm J W Sim
- Division of Intramural Research (DIR), Laboratory of Immunogenetics, NIAID, NIH, Bethesda, Maryland, USA
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | | | - Zachary Stotz
- Division of Intramural Research (DIR), Laboratory of Immunogenetics, NIAID, NIH, Bethesda, Maryland, USA
| | - Zhiya Yu
- Surgery Branch, NCI, NIH, Bethesda, Maryland, USA
| | - Jinghua Lu
- Division of Intramural Research (DIR), Laboratory of Immunogenetics, NIAID, NIH, Bethesda, Maryland, USA
| | - Paul Brennan
- Division of Intramural Research (DIR), Laboratory of Immunogenetics, NIAID, NIH, Bethesda, Maryland, USA
| | - Max Quastel
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - Geraldine M Gillespie
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - Eric O Long
- Division of Intramural Research (DIR), Laboratory of Immunogenetics, NIAID, NIH, Bethesda, Maryland, USA
| | - James C Yang
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - Peter D Sun
- Division of Intramural Research (DIR), Laboratory of Immunogenetics, NIAID, NIH, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Woessner NM, Brandl SM, Hartmann S, Schamel WW, Hartl FA, Minguet S. Phospho-mimetic CD3ε variants prevent TCR and CAR signaling. Front Immunol 2024; 15:1392933. [PMID: 38779683 PMCID: PMC11109380 DOI: 10.3389/fimmu.2024.1392933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction Antigen binding to the T cell antigen receptor (TCR) leads to the phosphorylation of the immunoreceptor tyrosine-based activation motifs (ITAMs) of the CD3 complex, and thereby to T cell activation. The CD3ε subunit plays a unique role in TCR activation by recruiting the kinase LCK and the adaptor protein NCK prior to ITAM phosphorylation. Here, we aimed to investigate how phosphorylation of the individual CD3ε ITAM tyrosines impacts the CD3ε signalosome. Methods We mimicked irreversible tyrosine phosphorylation by substituting glutamic acid for the tyrosine residues in the CD3ε ITAM. Results Integrating CD3ε phospho-mimetic variants into the complete TCR-CD3 complex resulted in reduced TCR signal transduction, which was partially compensated by the involvement of the other TCR-CD3 ITAMs. By using novel CD3ε phospho-mimetic Chimeric Antigen Receptor (CAR) variants, we avoided any compensatory effects of other ITAMs in the TCR-CD3 complex. We demonstrated that irreversible CD3ε phosphorylation prevented signal transduction upon CAR engagement. Mechanistically, we demonstrated that glutamic acid substitution at the N-terminal tyrosine residue of the CD3ε ITAM (Y39E) significantly reduces NCK binding to the TCR. In contrast, mutation at the C-terminal tyrosine of the CD3ε ITAM (Y50E) abolished LCK recruitment to the TCR, while increasing NCK binding. Double mutation at the C- and N-terminal tyrosines (Y39/50E) allowed ZAP70 to bind, but reduced the interaction with LCK and NCK. Conclusions The data demonstrate that the dynamic phosphorylation of the CD3ε ITAM tyrosines is essential for CD3ε to orchestrate optimal TCR and CAR signaling and highlights the key role of CD3ε signalosome to tune signal transduction.
Collapse
MESH Headings
- Humans
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- CD3 Complex/metabolism
- HEK293 Cells
- Immunoreceptor Tyrosine-Based Activation Motif
- Jurkat Cells
- Lymphocyte Activation/immunology
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics
- Phosphorylation
- Protein Binding
- Receptor-CD3 Complex, Antigen, T-Cell/metabolism
- Receptor-CD3 Complex, Antigen, T-Cell/immunology
- Receptor-CD3 Complex, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Signal Transduction/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- ZAP-70 Protein-Tyrosine Kinase/metabolism
- ZAP-70 Protein-Tyrosine Kinase/genetics
Collapse
Affiliation(s)
- Nadine M. Woessner
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Simon M. Brandl
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Sara Hartmann
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Wolfgang W. Schamel
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency (CCI), University Clinics and Medical Faculty, University, Freiburg, Germany
| | - Frederike A. Hartl
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency (CCI), University Clinics and Medical Faculty, University, Freiburg, Germany
| |
Collapse
|
4
|
Lee MS, Tuohy PJ, Kim CY, Yost PP, Lichauco K, Parrish HL, Van Doorslaer K, Kuhns MS. The CD4 transmembrane GGXXG and juxtamembrane (C/F)CV+C motifs mediate pMHCII-specific signaling independently of CD4-LCK interactions. eLife 2024; 12:RP88225. [PMID: 38639990 PMCID: PMC11031086 DOI: 10.7554/elife.88225] [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] [Indexed: 04/20/2024] Open
Abstract
CD4+ T cell activation is driven by five-module receptor complexes. The T cell receptor (TCR) is the receptor module that binds composite surfaces of peptide antigens embedded within MHCII molecules (pMHCII). It associates with three signaling modules (CD3γε, CD3δε, and CD3ζζ) to form TCR-CD3 complexes. CD4 is the coreceptor module. It reciprocally associates with TCR-CD3-pMHCII assemblies on the outside of a CD4+ T cells and with the Src kinase, LCK, on the inside. Previously, we reported that the CD4 transmembrane GGXXG and cytoplasmic juxtamembrane (C/F)CV+C motifs found in eutherian (placental mammal) CD4 have constituent residues that evolved under purifying selection (Lee et al., 2022). Expressing mutants of these motifs together in T cell hybridomas increased CD4-LCK association but reduced CD3ζ, ZAP70, and PLCγ1 phosphorylation levels, as well as IL-2 production, in response to agonist pMHCII. Because these mutants preferentially localized CD4-LCK pairs to non-raft membrane fractions, one explanation for our results was that they impaired proximal signaling by sequestering LCK away from TCR-CD3. An alternative hypothesis is that the mutations directly impacted signaling because the motifs normally play an LCK-independent role in signaling. The goal of this study was to discriminate between these possibilities. Using T cell hybridomas, our results indicate that: intracellular CD4-LCK interactions are not necessary for pMHCII-specific signal initiation; the GGXXG and (C/F)CV+C motifs are key determinants of CD4-mediated pMHCII-specific signal amplification; the GGXXG and (C/F)CV+C motifs exert their functions independently of direct CD4-LCK association. These data provide a mechanistic explanation for why residues within these motifs are under purifying selection in jawed vertebrates. The results are also important to consider for biomimetic engineering of synthetic receptors.
Collapse
Affiliation(s)
- Mark S Lee
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Peter J Tuohy
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Caleb Y Kim
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Philip P Yost
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Katrina Lichauco
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Heather L Parrish
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Koenraad Van Doorslaer
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
- School of Animal and Comparative Biomedical Sciences, The University of ArizonaTucsonUnited States
- Cancer Biology Graduate Interdisciplinary Program and Genetics Graduate Interdisciplinary Program, The University of ArizonaTucsonUnited States
- The BIO-5 Institute, The University of ArizonaTucsonUnited States
- The University of Arizona Cancer CenterTucsonUnited States
- The Arizona Center on Aging, The University of Arizona College of MedicineTucsonUnited States
| | - Michael S Kuhns
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
- Cancer Biology Graduate Interdisciplinary Program and Genetics Graduate Interdisciplinary Program, The University of ArizonaTucsonUnited States
- The BIO-5 Institute, The University of ArizonaTucsonUnited States
- The University of Arizona Cancer CenterTucsonUnited States
- The Arizona Center on Aging, The University of Arizona College of MedicineTucsonUnited States
| |
Collapse
|
5
|
Cochrane RW, Robino RA, Granger B, Allen E, Vaena S, Romeo MJ, de Cubas AA, Berto S, Ferreira LM. High affinity chimeric antigen receptor signaling induces an inflammatory program in human regulatory T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.31.587467. [PMID: 38617240 PMCID: PMC11014479 DOI: 10.1101/2024.03.31.587467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Regulatory T cells (Tregs) are promising cellular therapies to induce immune tolerance in organ transplantation and autoimmune disease. The success of chimeric antigen receptor (CAR) T-cell therapy for cancer has sparked interest in using CARs to generate antigen-specific Tregs. Here, we compared CAR with endogenous T cell receptor (TCR)/CD28 activation in human Tregs. Strikingly, CAR Tregs displayed increased cytotoxicity and diminished suppression of antigen-presenting cells and effector T (Teff) cells compared with TCR/CD28 activated Tregs. RNA sequencing revealed that CAR Tregs activate Teff cell gene programs. Indeed, CAR Tregs secreted high levels of inflammatory cytokines, with a subset of FOXP3+ CAR Tregs uniquely acquiring CD40L surface expression and producing IFNγ. Interestingly, decreasing CAR antigen affinity reduced Teff cell gene expression and inflammatory cytokine production by CAR Tregs. Our findings showcase the impact of engineered receptor activation on Treg biology and support tailoring CAR constructs to Tregs for maximal therapeutic efficacy.
Collapse
Affiliation(s)
- Russell W. Cochrane
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Rob A. Robino
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Bryan Granger
- Bioinformatics Core, Medical University of South Carolina, Charleston, SC, USA
| | - Eva Allen
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Silvia Vaena
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Martin J. Romeo
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Aguirre A. de Cubas
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Stefano Berto
- Bioinformatics Core, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Leonardo M.R. Ferreira
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
6
|
Jin Y, Yuan H, Mehta I, Ezenwa O, Morel PA. Alternatively Spliced Variants of Murine CD247 Influence T Cell Development and Activation, Revealing the Importance of the CD3ζ C-Terminal Region. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:541-550. [PMID: 38117282 PMCID: PMC10872740 DOI: 10.4049/jimmunol.2300511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023]
Abstract
CD247, also known as CD3ζ, is a crucial signaling molecule that transduces signals delivered by TCR through its three ITAMs. CD3ζ is required for successful thymocyte development. Three additional alternatively spliced variants of murine CD247 have been described, that is, CD3ι, CD3θ, and CD3η, that differ from CD3ζ in the C terminus such that the third ITAM is lost. Previous studies demonstrated defects in T cell development in mice expressing CD3η, but the TCR signaling pathways affected by CD3η and the impacts of the CD3ι and CD3θ on T cell development were not explored. In this study, we used a retrovirus-mediated gene transfer technique to express these three isoforms individually and examined the roles of them on T cell development and activation. Rag1-/- mice reconstituted with CD3θ-expressing bone marrow failed to develop mature T cells. CD3ι-expressing T cells exhibited similar development and activation as cells expressing CD3ζ. In contrast, thymic development was severely impaired in CD3η-reconstituted mice. Single-positive but not double-positive CD3η-expressing thymocytes had reduced TCR expression, and CD5 expression was decreased at the double-positive stage, suggesting a defect in positive selection. Peripheral CD3η-expressing T cells had expanded CD44hi populations and upregulation of exhaustion markers seen by flow cytometry and RNA sequencing analysis. Analysis of early signaling events demonstrated significantly reduced activation of both the PLCγ1 and Akt/mTOR signaling pathways. There was also a reduction in the frequency of activation of CD3η-expressing T cells. These studies reveal the importance of the CD3ζ C-terminal region in T cell development and activation.
Collapse
Affiliation(s)
- Ye Jin
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Huijuan Yuan
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh PA
| | - Isha Mehta
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ogechukwu Ezenwa
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Penelope A Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
7
|
Velasco Cárdenas RMH, Brandl SM, Meléndez AV, Schlaak AE, Buschky A, Peters T, Beier F, Serrels B, Taromi S, Raute K, Hauri S, Gstaiger M, Lassmann S, Huppa JB, Boerries M, Andrieux G, Bengsch B, Schamel WW, Minguet S. Harnessing CD3 diversity to optimize CAR T cells. Nat Immunol 2023; 24:2135-2149. [PMID: 37932456 PMCID: PMC10681901 DOI: 10.1038/s41590-023-01658-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 09/19/2023] [Indexed: 11/08/2023]
Abstract
Current US Food and Drug Administration-approved chimeric antigen receptor (CAR) T cells harbor the T cell receptor (TCR)-derived ζ chain as an intracellular activation domain in addition to costimulatory domains. The functionality in a CAR format of the other chains of the TCR complex, namely CD3δ, CD3ε and CD3γ, instead of ζ, remains unknown. In the present study, we have systematically engineered new CD3 CARs, each containing only one of the CD3 intracellular domains. We found that CARs containing CD3δ, CD3ε or CD3γ cytoplasmic tails outperformed the conventional ζ CAR T cells in vivo. Transcriptomic and proteomic analysis revealed differences in activation potential, metabolism and stimulation-induced T cell dysfunctionality that mechanistically explain the enhanced anti-tumor performance. Furthermore, dimerization of the CARs improved their overall functionality. Using these CARs as minimalistic and synthetic surrogate TCRs, we have identified the phosphatase SHP-1 as a new interaction partner of CD3δ that binds the CD3δ-ITAM on phosphorylation of its C-terminal tyrosine. SHP-1 attenuates and restrains activation signals and might thus prevent exhaustion and dysfunction. These new insights into T cell activation could promote the rational redesign of synthetic antigen receptors to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Rubí M-H Velasco Cárdenas
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Simon M Brandl
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Ana Valeria Meléndez
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Alexandra Emilia Schlaak
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Clinic for Internal Medicine II, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Annabelle Buschky
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Timo Peters
- Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, Vienna, Austria
| | - Fabian Beier
- Institute for Surgical Pathology, Medical Center, Freiburg, Germany
| | - Bryan Serrels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- NanoString Technologies, Inc., Seattle, WA, USA
| | - Sanaz Taromi
- Department of Medicine I, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Medical and Life Sciences, University of Furtwangen, Freiburg, Germany
| | - Katrin Raute
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Simon Hauri
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Silke Lassmann
- Institute for Surgical Pathology, Medical Center, Freiburg, Germany
| | - Johannes B Huppa
- Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, Vienna, Austria
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium and German Cancer Research Center, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bertram Bengsch
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Clinic for Internal Medicine II, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang W Schamel
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, Freiburg, Germany
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, Freiburg, Germany.
| |
Collapse
|
8
|
Yun K, Siegler EL, Kenderian SS. Who wins the combat, CAR or TCR? Leukemia 2023; 37:1953-1962. [PMID: 37626090 DOI: 10.1038/s41375-023-01976-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/04/2023] [Accepted: 07/17/2023] [Indexed: 08/27/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has drawn increasing attention over the last few decades given its remarkable effectiveness and breakthroughs in treating B cell hematological malignancies. Even though CAR-T cell therapy has outstanding clinical successes, most treated patients still relapse after infusion. CARs are derived from the T cell receptor (TCR) complex and co-stimulatory molecules associated with T cell activation; however, the similarities and differences between CARs and endogenous TCRs regarding their sensitivity, signaling pathway, killing mechanisms, and performance are still not fully understood. In this review, we discuss the parallel comparisons between CARs and TCRs from various aspects and how these current findings might provide novel insights and contribute to improvement of CAR-T cell therapy efficacy.
Collapse
Affiliation(s)
- Kun Yun
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Tanaka A, Maeda S, Nomura T, Llamas-Covarrubias MA, Tanaka S, Jin L, Lim EL, Morikawa H, Kitagawa Y, Akizuki S, Ito Y, Fujimori C, Hirota K, Murase T, Hashimoto M, Higo J, Zamoyska R, Ueda R, Standley DM, Sakaguchi N, Sakaguchi S. Construction of a T cell receptor signaling range for spontaneous development of autoimmune disease. J Exp Med 2023; 220:213728. [PMID: 36454183 PMCID: PMC9718937 DOI: 10.1084/jem.20220386] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/06/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
Thymic selection and peripheral activation of conventional T (Tconv) and regulatory T (Treg) cells depend on TCR signaling, whose anomalies are causative of autoimmunity. Here, we expressed in normal mice mutated ZAP-70 molecules with different affinities for the CD3 chains, or wild type ZAP-70 at graded expression levels under tetracycline-inducible control. Both manipulations reduced TCR signaling intensity to various extents and thereby rendered those normally deleted self-reactive thymocytes to become positively selected and form a highly autoimmune TCR repertoire. The signal reduction more profoundly affected Treg development and function because their TCR signaling was further attenuated by Foxp3 that physiologically repressed the expression of TCR-proximal signaling molecules, including ZAP-70, upon TCR stimulation. Consequently, the TCR signaling intensity reduced to a critical range generated pathogenic autoimmune Tconv cells and concurrently impaired Treg development/function, leading to spontaneous occurrence of autoimmune/inflammatory diseases, such as autoimmune arthritis and inflammatory bowel disease. These results provide a general model of how altered TCR signaling evokes autoimmune disease.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Frontier Research in Tumor Immunology, Center of Medical Innovation and Translational Research, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shinji Maeda
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Nomura
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mara Anais Llamas-Covarrubias
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Institute of Research in Biomedical Sciences, University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Satoshi Tanaka
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Lin Jin
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ee Lyn Lim
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Hiromasa Morikawa
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Yohko Kitagawa
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shuji Akizuki
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshinaga Ito
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Chihiro Fujimori
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Tosei Murase
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Motomu Hashimoto
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Junichi Higo
- Institute for Protein Research, Osaka University, Suita, Japan
| | - Rose Zamoyska
- Institute for Immunology and Infection Research, The University of Edinburgh, Edinburgh, UK
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Aichi, Japan
| | - Daron M Standley
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Noriko Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shimon Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
10
|
Goodman DB, Azimi CS, Kearns K, Talbot A, Garakani K, Garcia J, Patel N, Hwang B, Lee D, Park E, Vykunta VS, Shy BR, Ye CJ, Eyquem J, Marson A, Bluestone JA, Roybal KT. Pooled screening of CAR T cells identifies diverse immune signaling domains for next-generation immunotherapies. Sci Transl Med 2022; 14:eabm1463. [PMID: 36350984 PMCID: PMC9939256 DOI: 10.1126/scitranslmed.abm1463] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Chimeric antigen receptors (CARs) repurpose natural signaling components to retarget T cells to refractory cancers but have shown limited efficacy in persistent, recurrent malignancies. Here, we introduce "CAR Pooling," a multiplexed approach to rapidly identify CAR designs with clinical potential. Forty CARs with signaling domains derived from a range of immune cell lineages were evaluated in pooled assays for their ability to stimulate critical T cell effector functions during repetitive stimulation that mimics long-term tumor antigen exposure. Several domains were identified from the tumor necrosis factor (TNF) receptor family that have been primarily associated with B cells. CD40 enhanced proliferation, whereas B cell-activating factor receptor (BAFF-R) and transmembrane activator and CAML interactor (TACI) promoted cytotoxicity. These functions were enhanced relative to clinical benchmarks after prolonged antigen stimulation, and CAR T cell signaling through these domains fell into distinct states of memory, cytotoxicity, and metabolism. BAFF-R CAR T cells were enriched for a highly cytotoxic transcriptional signature previously associated with positive clinical outcomes. We also observed that replacing the 4-1BB intracellular signaling domain with the BAFF-R signaling domain in a clinically validated B cell maturation antigen (BCMA)-specific CAR resulted in enhanced activity in a xenotransplant model of multiple myeloma. Together, these results show that CAR Pooling is a general approach for rapid exploration of CAR architecture and activity to improve the efficacy of CAR T cell therapies.
Collapse
Affiliation(s)
- Daniel B. Goodman
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
- Gladstone UCSF Institute for Genetic Immunology; San Francisco, CA, 94107, USA
- School of Medicine, University of California, San Francisco; San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco; San Francisco, CA 94143, USA
| | - Camillia S. Azimi
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
| | - Kendall Kearns
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
| | - Alexis Talbot
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
- Gladstone UCSF Institute for Genetic Immunology; San Francisco, CA, 94107, USA
- INSERM U976, Saint Louis Research Institute, Paris City University, Paris, France
| | - Kiavash Garakani
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
| | - Julie Garcia
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
| | - Nisarg Patel
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco; San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco; San Francisco, CA, USA
- School of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Byungjin Hwang
- Institute for Human Genetics (IHG), University of California, San Francisco; San Francisco, California, USA
- Department of Medicine, University of California, San Francisco; San Francisco, California, 94143, USA
| | - David Lee
- Institute for Human Genetics (IHG), University of California, San Francisco; San Francisco, California, USA
- Department of Medicine, University of California, San Francisco; San Francisco, California, 94143, USA
| | - Emily Park
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
| | - Vivasvan S. Vykunta
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco; San Francisco, California, 94158, USA
- Gladstone UCSF Institute for Genetic Immunology; San Francisco, CA, 94107, USA
- School of Medicine, University of California, San Francisco; San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco; San Francisco, California, 94143, USA
| | - Brian R. Shy
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco; San Francisco, California, 94158, USA
- Gladstone UCSF Institute for Genetic Immunology; San Francisco, CA, 94107, USA
- School of Medicine, University of California, San Francisco; San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco; San Francisco, California, 94143, USA
| | - Chun Jimmie Ye
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
- Chan Zuckerberg Biohub; San Francisco, California, 94158, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco; San Francisco, CA, USA
- Institute for Human Genetics (IHG), University of California, San Francisco; San Francisco, California, USA
- Department of Epidemiology and Biostatistics, San Francisco; San Francisco, CA 94143, USA
- Department of Medicine, University of California, San Francisco; San Francisco, California, 94143, USA
| | - Justin Eyquem
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
- Gladstone UCSF Institute for Genetic Immunology; San Francisco, CA, 94107, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco; San Francisco, California, 94158, USA
- Chan Zuckerberg Biohub; San Francisco, California, 94158, USA
- Gladstone UCSF Institute for Genetic Immunology; San Francisco, CA, 94107, USA
- School of Medicine, University of California, San Francisco; San Francisco, CA, USA
- Institute for Human Genetics (IHG), University of California, San Francisco; San Francisco, California, USA
- Innovative Genomics Institute, University of California, Berkeley; Berkeley, CA 94720, USA
- Diabetes Center, University of California, San Francisco; San Francisco, CA 94143, USA
- Department of Medicine, University of California, San Francisco; San Francisco, California, 94143, USA
| | - Jeffrey A. Bluestone
- Diabetes Center, University of California, San Francisco; San Francisco, CA 94143, USA
- Sonoma Biotherapeutics; South San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco; San Francisco, California, 94143, USA
| | - Kole T. Roybal
- Department of Microbiology and Immunology, University of California, San Francisco; San Francisco, California, 94143, USA
- Parker Institute for Cancer Immunotherapy; San Francisco, California, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco; San Francisco, California, 94158, USA
- Chan Zuckerberg Biohub; San Francisco, California, 94158, USA
- Gladstone UCSF Institute for Genetic Immunology; San Francisco, CA, 94107, USA
- UCSF Cell Design Institute; San Francisco, California, 94158, USA
| |
Collapse
|
11
|
Gangopadhyay K, Roy A, Chandradasan AC, Roy S, Debnath O, SenGupta S, Chowdhury S, Das D, Das R. An evolutionary divergent thermodynamic brake in ZAP-70 fine-tunes the kinetic proofreading in T cells. J Biol Chem 2022; 298:102376. [PMID: 35970395 PMCID: PMC9486129 DOI: 10.1016/j.jbc.2022.102376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022] Open
Abstract
T cell signaling starts with assembling several tyrosine kinases and adaptor proteins to the T cell receptor (TCR), following the antigen-binding to the TCR. The stability of the TCR-antigen complex and the delay between the recruitment and activation of each kinase determines the T cell response. Integration of such delays constitutes a kinetic proofreading mechanism to regulate T cell response to the antigen binding. However, the mechanism of these delays is not fully understood. Combining biochemical experiments and kinetic modelling, here we report a thermodynamic brake in the regulatory module of the tyrosine kinase ZAP-70, which determines the ligand selectivity, and may delay the ZAP-70 activation upon antigen binding to TCR. The regulatory module of ZAP-70 comprises of a tandem SH2 (tSH2) domain that binds to its ligand, doubly-phosphorylated ITAM peptide (ITAM-Y2P), in two kinetic steps: a fast step and a slow step. We show the initial encounter complex formation between the ITAM-Y2P and tSH2 domain follows a fast-kinetic step, whereas the conformational transition to the holo-state follows a slow-kinetic step. We further observed a thermodynamic penalty imposed during the second phosphate-binding event reduces the rate of structural transition to the holo-state. Phylogenetic analysis revealed the evolution of the thermodynamic brake coincides with the divergence of the adaptive immune system to the cell-mediated and humoral responses. In addition, the paralogous kinase Syk expressed in B cells does not possess such a functional thermodynamic brake, which may explain the higher basal activation and lack of ligand selectivity in Syk.
Collapse
Affiliation(s)
- Kaustav Gangopadhyay
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Arnab Roy
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Athira C Chandradasan
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Swarnendu Roy
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Olivia Debnath
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Soumee SenGupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Subhankar Chowdhury
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Dipjyoti Das
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India.
| | - Rahul Das
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India; Centre for Advanced Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur, India.
| |
Collapse
|
12
|
Birtel M, Voss RH, Reinhard K, Rengstl B, Ouchan Y, Michel K, Hayduk N, Tillmann B, Becker R, Suchan M, Theobald M, Oehm P, Türeci Ö, Sahin U. A TCR-like CAR Promotes Sensitive Antigen Recognition and Controlled T-cell Expansion Upon mRNA Vaccination. CANCER RESEARCH COMMUNICATIONS 2022; 2:827-841. [PMID: 36923303 PMCID: PMC10010320 DOI: 10.1158/2767-9764.crc-21-0154] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/10/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022]
Abstract
Chimeric antigen receptor (CAR) T cells are efficacious in patients with B-cell malignancies, while their activity is limited in patients with solid tumors. We developed a novel heterodimeric TCR-like CAR (TCAR) designed to achieve optimal chain pairing and integration into the T-cell CD3 signaling complex. The TCAR mediated high antigen sensitivity and potent antigen-specific T-cell effector functions in short-term in vitro assays. Both persistence and functionality of TCAR T cells were augmented by provision of costimulatory signals, which improved proliferation in vitro and in vivo. Combination with a nanoparticulate RNA vaccine, developed for in vivo expansion of CAR T cells, promoted tightly controlled expansion, survival, and antitumor efficacy of TCAR T cells in vivo. Significance A novel TCAR is tightly controlled by RNA vaccine-mediated costimulation and may provide an alternative to second-generation CARs for the treatment of solid tumors.
Collapse
Affiliation(s)
- Matthias Birtel
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Ralf-Holger Voss
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
- Department of Research Center for Immunotherapy (FZI), University Medical Center (UMC) of the Johannes Gutenberg University, Mainz, Germany
| | - Katharina Reinhard
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Benjamin Rengstl
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Yasmina Ouchan
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Kristina Michel
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Nina Hayduk
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Bodo Tillmann
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
| | - René Becker
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
| | - Martin Suchan
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
| | - Matthias Theobald
- Department of Hematology, Oncology, and Pneumology, University Cancer Center (UCT), University Medical Center (UMC) of Johannes Gutenberg University, Mainz, Germany
| | - Petra Oehm
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Özlem Türeci
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Ugur Sahin
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
- Department of Research Center for Immunotherapy (FZI), University Medical Center (UMC) of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
13
|
Huseby ES, Teixeiro E. The perception and response of T cells to a changing environment are based on the law of initial value. Sci Signal 2022; 15:eabj9842. [PMID: 35639856 PMCID: PMC9290192 DOI: 10.1126/scisignal.abj9842] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
αβ T cells are critical components of the adaptive immune system and are capable of inducing sterilizing immunity after pathogen infection and eliminating transformed tumor cells. The development and function of T cells are controlled through the T cell antigen receptor, which recognizes peptides displayed on major histocompatibility complex (MHC) molecules. Here, we review how T cells generate the ability to recognize self-peptide-bound MHC molecules and use signals derived from these interactions to instruct cellular development, activation thresholds, and functional specialization in the steady state and during immune responses. We argue that the basic tenants of T cell development and function follow Weber-Fetcher's law of just noticeable differences and Wilder's law of initial value. Together, these laws argue that the ability of a system to respond and the quality of that response are scalable to the basal state of that system. Manifestation of these laws in T cells generates clone-specific activation thresholds that are based on perceivable differences between homeostasis and pathogen encounter (self versus nonself discrimination), as well as poised states for subsequent differentiation into specific effector cell lineages.
Collapse
Affiliation(s)
- Eric S. Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
14
|
Shinzawa M, Moseman EA, Gossa S, Mano Y, Bhattacharya A, Guinter T, Alag A, Chen X, Cam M, McGavern DB, Erman B, Singer A. Reversal of the T cell immune system reveals the molecular basis for T cell lineage fate determination in the thymus. Nat Immunol 2022; 23:731-742. [PMID: 35523960 PMCID: PMC9098387 DOI: 10.1038/s41590-022-01187-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/15/2022] [Indexed: 12/03/2022]
Abstract
T cell specificity and function are linked during development, as MHC-II-specific TCR signals generate CD4 helper T cells and MHC-I-specific TCR signals generate CD8 cytotoxic T cells, but the basis remains uncertain. We now report that switching coreceptor proteins encoded by Cd4 and Cd8 gene loci functionally reverses the T cell immune system, generating CD4 cytotoxic and CD8 helper T cells. Such functional reversal reveals that coreceptor proteins promote the helper-lineage fate when encoded by Cd4, but promote the cytotoxic-lineage fate when encoded in Cd8—regardless of the coreceptor proteins each locus encodes. Thus, T cell lineage fate is determined by cis-regulatory elements in coreceptor gene loci and is not determined by the coreceptor proteins they encode, invalidating coreceptor signal strength as the basis of lineage fate determination. Moreover, we consider that evolution selected the particular coreceptor proteins that Cd4 and Cd8 gene loci encode to avoid generating functionally reversed T cells because they fail to promote protective immunity against environmental pathogens. To determine how T cell lineage fates are determined in the thymus, Singer and colleagues generated ‘FlipFlop’ mice with a functionally reversed T cell immune system that distinguishes TCR signal strength versus TCR signal duration.
Collapse
Affiliation(s)
- Miho Shinzawa
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - E Ashley Moseman
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.,Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Selamawit Gossa
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yasuko Mano
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Abhisek Bhattacharya
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Terry Guinter
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amala Alag
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiongfong Chen
- Office of Science and Technology Resources, Office of the Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,CCR-SF Bioinformatics Group, Advanced Biomedical Computational Science, Biomedical Informatics and Data Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Maggie Cam
- Office of Science and Technology Resources, Office of the Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Batu Erman
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul, Turkey
| | - Alfred Singer
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
15
|
Wang H, Song X, Shen L, Wang X, Xu C. Exploiting T cell signaling to optimize engineered T cell therapies. Trends Cancer 2021; 8:123-134. [PMID: 34810156 DOI: 10.1016/j.trecan.2021.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 11/15/2022]
Abstract
Engineered T cell therapies, mainly chimeric antigen receptor (CAR)-T and T cell receptor (TCR)-T, have become the new frontier of cancer treatment. CAR-T and TCR-T therapies differ in many aspects, including cell persistence and toxicity, leading to different therapeutic outcomes. Both TCR and CAR recognize antigens and trigger T cell mediated antitumor response, but they have distinct molecular structures and signaling properties. TCR represents one of the most complex receptors, while CAR is a single-chain chimera integrating modules from multiple immune receptors. Understanding the mechanisms underlying the strengths and limitations of both systems can pave the way for the development of next-generation T cell therapy. This review synthesizes recent findings on TCR and CAR signaling and highlights the potential strategies of T cell engineering by signaling refinement.
Collapse
Affiliation(s)
- Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Shanghai Clinical Research and Trial Center, Shanghai, China.
| | - Xianming Song
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | - Chenqi Xu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| |
Collapse
|
16
|
Evnouchidou I, Caillens V, Koumantou D, Saveanu L. The role of endocytic trafficking in antigen T Cell Receptor activation. Biomed J 2021; 45:310-320. [PMID: 34592497 PMCID: PMC9250096 DOI: 10.1016/j.bj.2021.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/19/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022] Open
Abstract
Antigen T cell receptors (TCR) recognize antigenic peptides displayed by the major histocompatibility complex (pMHC) and play a critical role in T cell activation. The levels of TCR complexes at the cell surface, where signaling is initiated, depend on the balance between TCR synthesis, recycling and degradation. Cell surface TCR interaction with pMHC leads to receptor clustering and formation of a tight T cell-APC contact, the immune synapse, from which the activated TCR is internalized. While TCR internalization from the immune synapse has been initially considered to arrest TCR signaling, recent evidence support the hypothesis that the internalized receptor continues to signal from specialized endosomes. Here, we review the molecular mechanisms of TCR endocytosis and recycling, both in steady state and after T cell activation. We then discuss the experimental evidence in favor of endosomal TCR signaling and its possible consequences on T cell activation.
Collapse
Affiliation(s)
- Irini Evnouchidou
- Université de Paris, Centre de Recherche sur L'inflammation, INSERM U1149, CNRS ERL8252, Paris, France; Inovarion, Paris, France.
| | - Vivien Caillens
- Université de Paris, Centre de Recherche sur L'inflammation, INSERM U1149, CNRS ERL8252, Paris, France; Inovarion, Paris, France
| | - Despoina Koumantou
- Université de Paris, Centre de Recherche sur L'inflammation, INSERM U1149, CNRS ERL8252, Paris, France; Inovarion, Paris, France
| | - Loredana Saveanu
- Université de Paris, Centre de Recherche sur L'inflammation, INSERM U1149, CNRS ERL8252, Paris, France; Inovarion, Paris, France.
| |
Collapse
|
17
|
Wang X, Martin AD, Negri KR, McElvain ME, Oh J, Wu ML, Lee WH, Ando Y, Gabrelow GB, Toledo Warshaviak D, Sandberg ML, Xu H, Kamb A. Extensive functional comparisons between chimeric antigen receptors and T cell receptors highlight fundamental similarities. Mol Immunol 2021; 138:137-149. [PMID: 34419823 DOI: 10.1016/j.molimm.2021.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/13/2021] [Accepted: 07/21/2021] [Indexed: 12/22/2022]
Abstract
Though TCRs have been subject to limited engineering in the context of therapeutic design and optimization, they are used largely as found in nature. On the other hand, CARs are artificial, composed of different segments of proteins that function in the immune system. This characteristic raises the possibility of altered response to immune regulatory stimuli. Here we describe a large-scale, systematic comparison of CARs and TCRs across 5 different pMHC targets, with a total of 19 constructs examined in vitro. These functional measurements include CAR- and TCR-mediated activation, proliferation, and cytotoxicity in both acute and chronic settings. Surprisingly, we find no consistent difference between CARs and TCRs as receptor classes with respect to their relative sensitivity to major regulators of T cell activation: PD-L1, CD80/86 and IL-2. Though TCRs often emerge from human blood directly as potent, selective receptors, CARs must be heavily optimized to attain these properties for pMHC targets. Nonetheless, when iteratively improved and compared head to head in functional tests, CARs appear remarkably similar to TCRs with respect to immune modulation.
Collapse
Affiliation(s)
- Xueyin Wang
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Aaron D Martin
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Kathleen R Negri
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Michele E McElvain
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Julyun Oh
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Ming-Lun Wu
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Wen-Hua Lee
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Yuta Ando
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Grant B Gabrelow
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | | | - Mark L Sandberg
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States
| | - Han Xu
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States.
| | - Alexander Kamb
- A2 Biotherapeutics, 30301 Agoura Rd., Agoura Hills, CA, 91301, United States.
| |
Collapse
|
18
|
AlEjielat R, Khaleel A, Tarkhan AH. Differential gene expression analysis of ankylosing spondylitis shows deregulation of the HLA-DRB, HLA-DQB, ITM2A, and CTLA4 genes. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00161-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Ankylosing spondylitis (AS) is a rare inflammatory disorder affecting the spinal joints. Although we know some of the genetic factors that are associated with the disease, the molecular basis of this illness has not yet been fully elucidated, and the genes involved in AS pathogenesis have not been entirely identified. The current study aimed at constructing a gene network that may serve as an AS gene signature and biomarker, both of which will help in disease diagnosis and the identification of therapeutic targets. Previously published gene expression profiles of 16 AS patients and 16 gender- and age-matched controls that were profiled on the Illumina HumanHT-12 V3.0 Expression BeadChip platform were mined. Patients were Portuguese, 21 to 64 years old, were diagnosed based on the modified New York criteria, and had Bath Ankylosing Spondylitis Disease Activity Index scores > 4 and Bath Ankylosing Spondylitis Functional Index scores > 4. All patients were receiving only NSAIDs and/or sulphasalazine. Functional enrichment and pathway analysis were performed to create an interaction network of differentially expressed genes.
Results
ITM2A, ICOS, VSIG10L, CD59, TRAC, and CTLA-4 were among the significantly differentially expressed genes in AS, but the most significantly downregulated genes were the HLA-DRB6, HLA-DRB5, HLA-DRB4, HLA-DRB3, HLA-DRB1, HLA-DQB1, ITM2A, and CTLA-4 genes. The genes in this study were mostly associated with the regulation of the immune system processes, parts of cell membrane, and signaling related to T cell receptor and antigen receptor, in addition to some overlaps related to the IL2 STAT signaling, as well as the androgen response. The most significantly over-represented pathways in the data set were associated with the “RUNX1 and FOXP3 which control the development of regulatory T lymphocytes (Tregs)” and the “GABA receptor activation” pathways.
Conclusions
Comprehensive gene analysis of differentially expressed genes in AS reveals a significant gene network that is involved in a multitude of important immune and inflammatory pathways. These pathways and networks might serve as biomarkers for AS and can potentially help in diagnosing the disease and identifying future targets for treatment.
Collapse
|
19
|
Kent A, Longino NV, Christians A, Davila E. Naturally Occurring Genetic Alterations in Proximal TCR Signaling and Implications for Cancer Immunotherapy. Front Immunol 2021; 12:658611. [PMID: 34012443 PMCID: PMC8126620 DOI: 10.3389/fimmu.2021.658611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
T cell-based immunotherapies including genetically engineered T cells, adoptive transfer of tumor-infiltrating lymphocytes, and immune checkpoint blockade highlight the impressive anti-tumor effects of T cells. These successes have provided new hope to many cancer patients with otherwise poor prognoses. However, only a fraction of patients demonstrates durable responses to these forms of therapies and many develop significant immune-mediated toxicity. These heterogeneous clinical responses suggest that underlying nuances in T cell genetics, phenotypes, and activation states likely modulate the therapeutic impact of these approaches. To better characterize known genetic variations that may impact T cell function, we 1) review the function of early T cell receptor-specific signaling mediators, 2) offer a synopsis of known mutations and genetic alterations within the associated molecules, 3) discuss the link between these mutations and human disease and 4) review therapeutic strategies under development or in clinical testing that target each of these molecules for enhancing anti-tumor T cell activity. Finally, we discuss novel engineering approaches that could be designed based on our understanding of the function of these molecules in health and disease.
Collapse
Affiliation(s)
- Andrew Kent
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
| | - Natalie V. Longino
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Allison Christians
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
| | - Eduardo Davila
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
20
|
Rudd CE. How the Discovery of the CD4/CD8-p56 lck Complexes Changed Immunology and Immunotherapy. Front Cell Dev Biol 2021; 9:626095. [PMID: 33791292 PMCID: PMC8005572 DOI: 10.3389/fcell.2021.626095] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/11/2021] [Indexed: 12/22/2022] Open
Abstract
The past 25 years have seen enormous progress in uncovering the receptors and signaling mechanisms on T-cells that activate their various effecter functions. Until the late 1980s, most studies on T-cells had focused on the influx of calcium and the levels of cAMP/GMP in T-cells. My laboratory then uncovered the interaction of CD4 and CD8 co-receptors with the protein-tyrosine kinase p56lck which are now widely accepted as the initiators of the tyrosine phosphorylation cascade leading to T-cell activation. The finding explained how immune recognition receptors expressed by many immune cells, which lack intrinsic catalytic activity, can transduce activation signals via non-covalent association with non-receptor tyrosine kinases. The discovery also established the concept that a protein tyrosine phosphorylation cascade operated in T-cells. In this vein, we and others then showed that the CD4- and CD8-p56lck complexes phosphorylate the TCR complexes which led to the identification of other protein-tyrosine kinases such as ZAP-70 and an array of substrates that are now central to studies in T-cell immunity. Other receptors such as B-cell receptor, Fc receptors and others were also subsequently found to use src kinases to control cell growth. In T-cells, p56lck driven phosphorylation targets include co-receptors such as CD28 and CTLA-4 and immune cell-specific adaptor proteins such as LAT and SLP-76 which act to integrate signals proximal to surface receptors. CD4/CD8-p56lck regulated events in T-cells include intracellular calcium mobilization, integrin activation and the induction of transcription factors for gene expression. Lastly, the identification of the targets of p56lck in the TCR and CD28 provided the framework for the development of chimeric antigen receptor (CAR) therapy in the treatment of cancer. In this review, I outline a history of the development of events that led to the development of the "TCR signaling paradigm" and its implications to immunology and immunotherapy.
Collapse
Affiliation(s)
- Christopher E. Rudd
- Division of Immunology-Oncology, Centre de Recherche Hôpital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Faculty of Medicine, Universite de Montreal, Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University Health Center, McGill University, Montreal, QC, Canada
| |
Collapse
|
21
|
Boddul SV, Sharma RK, Dubnovitsky A, Raposo B, Gerstner C, Shen Y, Iyer VS, Kasza Z, Kwok WW, Winkler AR, Klareskog L, Malmström V, Bettini M, Wermeling F. In vitro and ex vitro functional characterization of human HLA-DRB1∗04 restricted T cell receptors. J Transl Autoimmun 2021; 4:100087. [PMID: 33768201 PMCID: PMC7980064 DOI: 10.1016/j.jtauto.2021.100087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Recent advances in single-cell sequencing technologies enable the generation of large-scale data sets of paired TCR sequences from patients with autoimmune disease. Methods to validate and characterize patient-derived TCR data are needed, as well as relevant model systems that can support the development of antigen-specific tolerance inducing drugs. We have generated a pipeline to allow streamlined generation of 'artificial' T cells in a robust and reasonably high throughput manner for in vitro and in vivo studies of antigen-specific and patient-derived immune responses. Hereby chimeric (mouse-human) TCR alpha and beta constructs are re-expressed in three different formats for further studies: (i) transiently in HEK cells for peptide-HLA tetramer validation experiments, (ii) stably in the TCR-negative 58 T cell line for functional readouts such as IL-2 production and NFAT-signaling, and lastly (iii) in human HLA-transgenic mice for studies of autoimmune disease and therapeutic interventions. As a proof of concept, we have used human HLA-DRB1∗04:01 restricted TCR sequences specific for a type I diabetes-associated GAD peptide, and an influenza-derived HA peptide. We show that the same chimeric TCR constructs can be used in each of the described assays facilitating sequential validation and prioritization steps leading to humanized animal models.
Collapse
Key Words
- APC, antigen presenting cells
- BM, bone marrow
- Ca2+, calcium
- Cell lines
- GAD, glutamic acid decarboxylase
- GFP, green fluorescent protein
- GWAS, Genome-wide association studies
- HA, Influenza hemagglutinin
- HLA
- HLA, Human leukocyte antigen
- HSCs, hematopoietic stem cells
- Humanized animal models
- MHC, major histocompatibility complex
- NFAT, Nuclear factor of activated T-cells
- RA, Rheumatoid arthritis
- RAG, Recombination-activating genes
- T1D, Type-1 diabetes
- TCR
- TCR, T cell receptor
- TCRa, TCR alpha
- TCRb, TCR beta
- TMR, HLA tetramer
- Tolerance
- hCD4, human CD4
- hTCR, human TCR
Collapse
Affiliation(s)
- Sanjaykumar V Boddul
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Ravi Kumar Sharma
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Anatoly Dubnovitsky
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Bruno Raposo
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Christina Gerstner
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Yunbing Shen
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Vaishnavi Srinivasan Iyer
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden.,School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore
| | - Zsolt Kasza
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - William W Kwok
- Translational Research Program, BRI at Virginia Mason, Seattle, WA, USA
| | - Aaron R Winkler
- Department of Inflammation and Immunology, Pfizer Inc., Cambridge, MA, USA
| | - Lars Klareskog
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Maria Bettini
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Fredrik Wermeling
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Trefzer A, Kadam P, Wang SH, Pennavaria S, Lober B, Akçabozan B, Kranich J, Brocker T, Nakano N, Irmler M, Beckers J, Straub T, Obst R. Dynamic adoption of anergy by antigen-exhausted CD4 + T cells. Cell Rep 2021; 34:108748. [PMID: 33567282 DOI: 10.1016/j.celrep.2021.108748] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 09/21/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Exhausted immune responses to chronic diseases represent a major challenge to global health. We study CD4+ T cells in a mouse model with regulatable antigen presentation. When the cells are driven through the effector phase and are then exposed to different levels of persistent antigen, they lose their T helper 1 (Th1) functions, upregulate exhaustion markers, resemble naturally anergic cells, and modulate their MAPK, mTORC1, and Ca2+/calcineurin signaling pathways with increasing dose and time. They also become unable to help B cells and, at the highest dose, undergo apoptosis. Transcriptomic analyses show the dynamic adjustment of gene expression and the accumulation of T cell receptor (TCR) signals over a period of weeks. Upon antigen removal, the cells recover their functionality while losing exhaustion and anergy markers. Our data suggest an adjustable response of CD4+ T cells to different levels of persisting antigen and contribute to a better understanding of chronic disease.
Collapse
Affiliation(s)
- Anne Trefzer
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Pallavi Kadam
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Shu-Hung Wang
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Stefanie Pennavaria
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Benedikt Lober
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Batuhan Akçabozan
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Jan Kranich
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Thomas Brocker
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Naoko Nakano
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, 85764 Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, 85764 Neuherberg, Germany; Chair of Experimental Genetics, Technische Universität München, 85354 Freising, Germany; German Center for Diabetes Research (DZD e. V.), Neuherberg, Germany
| | - Tobias Straub
- Bioinformatics Core Facility, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Reinhard Obst
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany.
| |
Collapse
|
23
|
Landry LG, Mann SE, Anderson AM, Nakayama M. Multiplex T-cell Stimulation Assay Utilizing a T-cell Activation Reporter-based Detection System. Bio Protoc 2021; 11:e3883. [PMID: 33732772 DOI: 10.21769/bioprotoc.3883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 01/10/2023] Open
Abstract
Immune tolerance and response are both largely driven by the interactions between the major histocompatibility complex (MHC) expressed by antigen presenting cells (APCs), T-cell receptors (TCRs) on T-cells, and their cognate antigens. Disordered interactions cause the pathogenesis of autoimmune diseases such as type 1 diabetes. Therefore, the identification of antigenic epitopes of autoreactive T-cells leads to important advances in therapeutics and biomarkers. Next-generation sequencing methods allow for the rapid identification of thousands of TCR clonotypes from single T-cells, and thus there is a need to determine cognate antigens for identified TCRs. This protocol describes a reporter system of T-cell activation where the fluorescent reporter protein ZsGreen-1 is driven by nuclear factor of activated T-cells (NFAT) signaling and read by flow cytometry. Reporter T-cells also constitutively express additional pairs of fluorescent proteins as identifiers, allowing for multiplexing of up to eight different reporter T-cell lines simultaneously, each expressing a different TCR of interest and distinguishable by flow cytometry. Once TCR expression cell lines are made they can be used indefinitely for making new T-cell lines with just one transduction step. This multiplexing system permits screening numbers of TCR-antigen interactions that would otherwise be impractical, can be used in a variety of contexts (i.e., screening individual antigens or antigen pools), and can be applied to study any T-cell-MHC-antigen trimolecular interaction.
Collapse
Affiliation(s)
- Laurie G Landry
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
| | - Sarah E Mann
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
| | - Amanda M Anderson
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
24
|
A biomimetic five-module chimeric antigen receptor ( 5MCAR) designed to target and eliminate antigen-specific T cells. Proc Natl Acad Sci U S A 2020; 117:28950-28959. [PMID: 33139567 DOI: 10.1073/pnas.2012495117] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
T cells express clonotypic T cell receptors (TCRs) that recognize peptide antigens in the context of class I or II MHC molecules (pMHCI/II). These receptor modules associate with three signaling modules (CD3γε, δε, and ζζ) and work in concert with a coreceptor module (either CD8 or CD4) to drive T cell activation in response to pMHCI/II. Here, we describe a first-generation biomimetic five-module chimeric antigen receptor (5MCAR). We show that 1) chimeric receptor modules built with the ectodomains of pMHCII assemble with CD3 signaling modules into complexes that redirect cytotoxic T lymphocyte (CTL) specificity and function in response to the clonotypic TCRs of pMHCII-specific CD4+ T cells, and 2) surrogate coreceptor modules enhance the function of these complexes. Furthermore, we demonstrate that adoptively transferred 5MCAR-CTLs can mitigate type I diabetes by targeting autoimmune CD4+ T cells in NOD mice. This work provides a framework for the construction of biomimetic 5MCARs that can be used as tools to study the impact of particular antigen-specific T cells in immune responses, and may hold potential for ameliorating diseases mediated by pathogenic T cells.
Collapse
|
25
|
Chandler NJ, Call MJ, Call ME. T Cell Activation Machinery: Form and Function in Natural and Engineered Immune Receptors. Int J Mol Sci 2020; 21:E7424. [PMID: 33050044 PMCID: PMC7582382 DOI: 10.3390/ijms21197424] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
The impressive success of chimeric antigen receptor (CAR)-T cell therapies in treating advanced B-cell malignancies has spurred a frenzy of activity aimed at developing CAR-T therapies for other cancers, particularly solid tumors, and optimizing engineered T cells for maximum clinical benefit in many different disease contexts. A rapidly growing body of design work is examining every modular component of traditional single-chain CARs as well as expanding out into many new and innovative engineered immunoreceptor designs that depart from this template. New approaches to immune cell and receptor engineering are being reported with rapidly increasing frequency, and many recent high-quality reviews (including one in this special issue) provide comprehensive coverage of the history and current state of the art in CAR-T and related cellular immunotherapies. In this review, we step back to examine our current understanding of the structure-function relationships in natural and engineered lymphocyte-activating receptors, with an eye towards evaluating how well the current-generation CAR designs recapitulate the most desirable features of their natural counterparts. We identify key areas that we believe are under-studied and therefore represent opportunities to further improve our grasp of form and function in natural and engineered receptors and to rationally design better therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Chandler
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Melissa J. Call
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Matthew E. Call
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
26
|
Wu W, Zhou Q, Masubuchi T, Shi X, Li H, Xu X, Huang M, Meng L, He X, Zhu H, Gao S, Zhang N, Jing R, Sun J, Wang H, Hui E, Wong CC, Xu C. Multiple Signaling Roles of CD3ε and Its Application in CAR-T Cell Therapy. Cell 2020; 182:855-871.e23. [PMID: 32730808 DOI: 10.1016/j.cell.2020.07.018] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/28/2020] [Accepted: 07/14/2020] [Indexed: 01/17/2023]
Abstract
A T cell receptor (TCR) mediates antigen-induced signaling through its associated CD3ε, δ, γ, and ζ, but the contributions of different CD3 chains remain elusive. Using quantitative mass spectrometry, we simultaneously quantitated the phosphorylation of the immunoreceptor tyrosine-based activation motif (ITAM) of all CD3 chains upon TCR stimulation. A subpopulation of CD3ε ITAMs was mono-phosphorylated, owing to Lck kinase selectivity, and specifically recruited the inhibitory Csk kinase to attenuate TCR signaling, suggesting that TCR is a self-restrained signaling machinery containing both activating and inhibitory motifs. Moreover, we found that incorporation of the CD3ε cytoplasmic domain into a second-generation chimeric antigen receptor (CAR) improved antitumor activity of CAR-T cells. Mechanistically, the Csk-recruiting ITAM of CD3ε reduced CAR-T cytokine production whereas the basic residue rich sequence (BRS) of CD3ε promoted CAR-T persistence via p85 recruitment. Collectively, CD3ε is a built-in multifunctional signal tuner, and increasing CD3 diversity represents a strategy to design next-generation CAR.
Collapse
Affiliation(s)
- Wei Wu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiuping Zhou
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Takeya Masubuchi
- Section of Cell & Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0347, USA
| | - Xiaoshan Shi
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Center for Precision Medicine Multi-omics Research, Peking University Health Science Center, Peking University First Hospital, Beijing 100191, China
| | - Hua Li
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyi Xu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Min Huang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Meng
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xing He
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hengyu Zhu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuaixin Gao
- Center for Precision Medicine Multi-omics Research, Peking University Health Science Center, Peking University First Hospital, Beijing 100191, China
| | - Nan Zhang
- Center for Precision Medicine Multi-omics Research, Peking University Health Science Center, Peking University First Hospital, Beijing 100191, China
| | - Ruirui Jing
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Institute of Hematology, Zhejiang University & Laboratory of Stem Cell and Immunotherapy Engineering, Hangzhou 310058, Zhejiang, China
| | - Jie Sun
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Institute of Hematology, Zhejiang University & Laboratory of Stem Cell and Immunotherapy Engineering, Hangzhou 310058, Zhejiang, China
| | - Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Enfu Hui
- Section of Cell & Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0347, USA.
| | - Catherine Chiulan Wong
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Center for Precision Medicine Multi-omics Research, Peking University Health Science Center, Peking University First Hospital, Beijing 100191, China; School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| | - Chenqi Xu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, Zhejiang, China.
| |
Collapse
|
27
|
Meng X, Jing R, Qian L, Zhou C, Sun J. Engineering Cytoplasmic Signaling of CD28ζ CARs for Improved Therapeutic Functions. Front Immunol 2020; 11:1046. [PMID: 32636832 PMCID: PMC7318076 DOI: 10.3389/fimmu.2020.01046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/30/2020] [Indexed: 12/29/2022] Open
Abstract
Chimeric antigen receptor modified T cells (CAR-T) have yielded impressive clinical outcomes in treating hematopoietic malignancies. However, relapses have occurred in a substantial number of patients and limited the development of CAR-T therapy. Most underlying reasons for these relapses can be attributed to poor persistence and rapid exhaustion of CAR-T cells in vivo. Despite multiple strategies having been developed, how to improve CAR-T persistence or resist exhaustion while maintaining sufficient cytotoxic functions is still a great challenge. Here we discuss engineering cytoplasmic signaling as an important strategy for CAR optimization. This review summarizes recent advances showing that the anti-tumor function of CAR-T cells can be improved by optimizing the CD3ζ domain or downstream signaling of CD28ζ CAR.
Collapse
Affiliation(s)
- Xianhui Meng
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
- Institute of Hematology, Zhejiang University & Laboratory of Stem Cell and Immunotherapy Engineering, Hangzhou, China
| | - Ruirui Jing
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
- Institute of Hematology, Zhejiang University & Laboratory of Stem Cell and Immunotherapy Engineering, Hangzhou, China
| | - Liling Qian
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
- Institute of Hematology, Zhejiang University & Laboratory of Stem Cell and Immunotherapy Engineering, Hangzhou, China
| | - Chun Zhou
- School of Public Health and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Sun
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
- Institute of Hematology, Zhejiang University & Laboratory of Stem Cell and Immunotherapy Engineering, Hangzhou, China
| |
Collapse
|
28
|
Dunst J, Glaros V, Englmaier L, Sandoz PA, Önfelt B, Kisielow J, Kreslavsky T. Recognition of synthetic polyanionic ligands underlies "spontaneous" reactivity of Vγ1 γδTCRs. J Leukoc Biol 2020; 107:1033-1044. [PMID: 31943366 PMCID: PMC7317387 DOI: 10.1002/jlb.2ma1219-392r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/05/2019] [Accepted: 12/11/2019] [Indexed: 01/02/2023] Open
Abstract
Although γδTCRs were discovered more than 30 yr ago, principles of antigen recognition by these receptors remain unclear and the nature of these antigens is largely elusive. Numerous studies reported that T cell hybridomas expressing several Vγ1-containing TCRs, including the Vγ1Vδ6 TCR of γδNKT cells, spontaneously secrete cytokines. This property was interpreted as recognition of a self-ligand expressed on the hybridoma cells themselves. Here, we revisited this finding using a recently developed reporter system and live single cell imaging. We confirmed strong spontaneous signaling by Vγ1Vδ6 and related TCRs, but not by TCRs from several other γδ or innate-like αβ T cells, and demonstrated that both γ and δ chains contributed to this reactivity. Unexpectedly, live single cell imaging showed that activation of this signaling did not require any interaction between cells. Further investigation revealed that the signaling is instead activated by interaction with negatively charged surfaces abundantly present under regular cell culture conditions and was abrogated when noncharged cell culture vessels were used. This mode of TCR signaling activation was not restricted to the reporter cell lines, as interaction with negatively charged surfaces also triggered TCR signaling in ex vivo Vγ1 γδ T cells. Taken together, these results explain long-standing observations on the spontaneous reactivity of Vγ1Vδ6 TCR and demonstrate an unexpected antigen presentation-independent mode of TCR activation by a spectrum of chemically unrelated polyanionic ligands.
Collapse
Affiliation(s)
- Josefine Dunst
- Department of Medicine, Division of Immunology and Allergy, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Vassilis Glaros
- Department of Medicine, Division of Immunology and Allergy, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Lukas Englmaier
- Department of Medicine, Division of Immunology and Allergy, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Patrick A. Sandoz
- Department of Applied PhysicsScience for Life LaboratoryKTH Royal Institute of TechnologyStockholmSweden
| | - Björn Önfelt
- Department of Applied PhysicsScience for Life LaboratoryKTH Royal Institute of TechnologyStockholmSweden
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstituteSolnaSweden
| | - Jan Kisielow
- Institute of Molecular Health SciencesETHZurichSwitzerland
| | - Taras Kreslavsky
- Department of Medicine, Division of Immunology and Allergy, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
29
|
White J, O'Brien RL, Born WK. BW5147 and Derivatives for the Study of T Cells and their Antigen Receptors. Arch Immunol Ther Exp (Warsz) 2020; 68:15. [PMID: 32419056 DOI: 10.1007/s00005-020-00579-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/30/2020] [Indexed: 10/24/2022]
Abstract
Like B cells, T cells can be immortalized through hybridization with lymphoma cells, a technique that has been particularly useful in the study of the T cell receptors (TCR) for antigen. In T cell hybridizations, the AKR mouse strain-derived thymus lymphoma BW5147 is by far the most popular fusion line. However, the full potential of this technology had to await inactivation of the productively rearranged TCR-α and -β genes in the lymphoma. BWα-β-, the TCR-gene deficient variant of the original lymphoma, which has become the fusion line of choice for αβ T cells, is now available with numerous modifications, enabling the investigation of many aspects of TCR-mediated responses and TCR-structure. Unexpectedly, inactivating BW's functional TCR-α gene also rendered the lymphoma more permissive for the expression of TCR-γδ, facilitating the study of γδ T cells, their TCRs, and their TCR-mediated reactivity.
Collapse
Affiliation(s)
- Janice White
- Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Rebecca L O'Brien
- Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA.,Department of Immunology and Microbiology, University of Colorado Health Sciences Center, Aurora, CO, 80045, USA
| | - Willi K Born
- Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA. .,Department of Immunology and Microbiology, University of Colorado Health Sciences Center, Aurora, CO, 80045, USA.
| |
Collapse
|
30
|
Alpaca ( Vicugna pacos), the first nonprimate species with a phosphoantigen-reactive Vγ9Vδ2 T cell subset. Proc Natl Acad Sci U S A 2020; 117:6697-6707. [PMID: 32139608 DOI: 10.1073/pnas.1909474117] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Vγ9Vδ2 T cells are a major γδ T cell population in the human blood expressing a characteristic Vγ9JP rearrangement paired with Vδ2. This cell subset is activated in a TCR-dependent and MHC-unrestricted fashion by so-called phosphoantigens (PAgs). PAgs can be microbial [(E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate, HMBPP] or endogenous (isopentenyl pyrophosphate, IPP) and PAg sensing depends on the expression of B7-like butyrophilin (BTN3A, CD277) molecules. IPP increases in some transformed or aminobisphosphonate-treated cells, rendering those cells a target for Vγ9Vδ2 T cells in immunotherapy. Yet, functional Vγ9Vδ2 T cells have only been described in humans and higher primates. Using a genome-based study, we showed in silico translatable genes encoding Vγ9, Vδ2, and BTN3 in a few nonprimate mammalian species. Here, with the help of new monoclonal antibodies, we directly identified a T cell population in the alpaca (Vicugna pacos), which responds to PAgs in a BTN3-dependent fashion and shows typical TRGV9- and TRDV2-like rearrangements. T cell receptor (TCR) transductants and BTN3-deficient human 293T cells reconstituted with alpaca or human BTN3 or alpaca/human BTN3 chimeras showed that alpaca Vγ9Vδ2 TCRs recognize PAg in the context of human and alpaca BTN3. Furthermore, alpaca BTN3 mediates PAg recognition much better than human BTN3A1 alone and this improved functionality mapped to the transmembrane/cytoplasmic part of alpaca BTN3. In summary, we found remarkable similarities but also instructive differences of PAg-recognition by human and alpaca, which help in better understanding the molecular mechanisms controlling the activation of this prominent population of γδ T cells.
Collapse
|
31
|
Schamel WW, Alarcon B, Minguet S. The TCR is an allosterically regulated macromolecular machinery changing its conformation while working. Immunol Rev 2020; 291:8-25. [PMID: 31402501 DOI: 10.1111/imr.12788] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 12/13/2022]
Abstract
The αβ T-cell receptor (TCR) is a multiprotein complex controlling the activation of T cells. Although the structure of the complete TCR is not known, cumulative evidence supports that the TCR cycles between different conformational states that are promoted either by thermal motion or by force. These structural transitions determine whether the TCR engages intracellular effectors or not, regulating TCR phosphorylation and signaling. As for other membrane receptors, ligand binding selects and stabilizes the TCR in active conformations, and/or switches the TCR to activating states that were not visited before ligand engagement. Here we review the main models of TCR allostery, that is, ligand binding at TCRαβ changes the structure at CD3 and ζ. (a) The ITAM and proline-rich sequence exposure model, in which the TCR's cytoplasmic tails shield each other and ligand binding exposes them for phosphorylation. (b) The membrane-ITAM model, in which the CD3ε and ζ tails are sequestered inside the membrane and again ligand binding exposes them. (c) The mechanosensor model in which ligand binding exerts force on the TCR, inducing structural changes that allow signaling. Since these models are complementary rather than competing, we propose a unified model that aims to incorporate all existing data.
Collapse
Affiliation(s)
- Wolfgang W Schamel
- Department of Immunology, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Balbino Alarcon
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | - Susana Minguet
- Department of Immunology, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| |
Collapse
|
32
|
Takeuchi Y, Hirota K, Sakaguchi S. Impaired T cell receptor signaling and development of T cell-mediated autoimmune arthritis. Immunol Rev 2020; 294:164-176. [PMID: 31944330 DOI: 10.1111/imr.12841] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022]
Abstract
Mutations of the genes encoding T-cell receptor (TCR)-proximal signaling molecules, such as ZAP-70, can be causative of immunological diseases ranging from T-cell immunodeficiency to T-cell-mediated autoimmune disease. For example, SKG mice, which carry a hypomorphic point mutation of the Zap-70 gene, spontaneously develop T-cell-mediated autoimmune arthritis immunopathologically similar to human rheumatoid arthritis (RA). The Zap-70 mutation alters the sensitivity of developing T cells to thymic positive/negative selection by self-peptides/MHC complexes, shifting self-reactive TCR repertoire to include a dominant arthritogenic specificity and also affecting thymic development and function of autoimmune suppressive regulatory T (Treg) cells. Polyclonal self-reactive T cells, including potentially arthritogenic T cells, thus produced by the thymus recognize self-peptide/MHC complexes on antigen-presenting cells (APCs) in the periphery and stimulate them to produce cytokines including IL-6 to drive the arthritogenic T cells to differentiate into arthritogenic T-helper 17 (Th17) cells. Insufficient Treg suppression or activation of APCs via microbial and other environmental stimuli evokes arthritis by activating granulocyte-macrophage colony-stimulating factor-secreting effector Th17 cells, mediating chronic bone-destructive joint inflammation by activating myeloid cells, innate lymphoid cells, and synoviocytes in the joint. These findings obtained from the study of SKG mouse arthritis are instrumental in understanding how arthritogenic T cells are produced, become activated, and differentiate into effector T cells mediating arthritis, and may help devising therapeutic measures targeting autoimmune pathogenic Th17 cells or autoimmune-suppressing Treg cells to treat and prevent RA.
Collapse
Affiliation(s)
- Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
33
|
Abstract
Advances in academic and clinical studies during the last several years have resulted in practical outcomes in adoptive immune therapy of cancer. Immune cells can be programmed with molecular modules that increase their therapeutic potency and specificity. It has become obvious that successful immunotherapy must take into account the full complexity of the immune system and, when possible, include the use of multifactor cell reprogramming that allows fast adjustment during the treatment. Today, practically all immune cells can be stably or transiently reprogrammed against cancer. Here, we review works related to T cell reprogramming, as the most developed field in immunotherapy. We discuss factors that determine the specific roles of αβ and γδ T cells in the immune system and the structure and function of T cell receptors in relation to other structures involved in T cell target recognition and immune response. We also discuss the aspects of T cell engineering, specifically the construction of synthetic T cell receptors (synTCRs) and chimeric antigen receptors (CARs) and the use of engineered T cells in integrative multifactor therapy of cancer.
Collapse
Affiliation(s)
- Samuel G Katz
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
34
|
Green EW, Bunse L, Bozza M, Sanghvi K, Platten M. TCR validation toward gene therapy for cancer. Methods Enzymol 2019; 629:419-441. [PMID: 31727252 DOI: 10.1016/bs.mie.2019.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The speed of T cell receptor (TCR) discovery has been revolutionized by barcode-based TCR sequencing approaches that allow the reconstitution of a T cell's paired alpha and beta TCR chain, and the process of TCR discovery promises to become ever faster and cheaper with the continuing development single cell analysis techniques. This technological progress has generated an urgent need to develop efficient TCR validation platforms for the rapid and safe clinical translation of TCRs into therapeutic agents. Whereas much attention has in the past focused on CD8-positive cytotoxic T cells recognizing MHC class I presented epitopes, the increasing demand to validate TCRs expressed on neoepitope-reactive CD4 T cells requires the implementation of large-scale T cell activation-based readout assays to complement existing multimer and cytotoxicity-based assays. Here, we present commonly used TCR validation assays, and include detailed guidance on TCR synthesis, delivery, and appropriate experimental control TCRs. We also comment on upcoming methods that hold promise for further speeding the process of TCR validation, hastening the translation of TCRs from the laboratory into the clinic.
Collapse
Affiliation(s)
- Edward W Green
- German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Lukas Bunse
- German Cancer Research Center, DKFZ, Heidelberg, Germany; University Hospital Mannheim, Mannheim, Germany; University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Bozza
- German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Khwab Sanghvi
- German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Michael Platten
- German Cancer Research Center, DKFZ, Heidelberg, Germany; University Hospital Mannheim, Mannheim, Germany; University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
35
|
Yamaguchi T, Teraguchi S, Furusawa C, Machiyama H, Watanabe TM, Fujita H, Sakaguchi S, Yanagida T. Theoretical modeling reveals that regulatory T cells increase T-cell interaction with antigen-presenting cells for stable immune tolerance. Int Immunol 2019; 31:743-753. [PMID: 31131864 PMCID: PMC6794947 DOI: 10.1093/intimm/dxz043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/24/2019] [Indexed: 01/22/2023] Open
Abstract
The immune system in tolerance maintains cell diversity without responding to self-antigens. Foxp3-expressing CD25+CD4+ regulatory T cells (Tregs) inhibit T-cell activation through various molecular mechanisms. However, several key questions are still not resolved, including how Tregs control the immune response on the basis of their self-skewed T-cell receptor repertoire and how Tregs avoid impeding relevant immunity against pathogens. Here, we show that Tregs promote the proliferation of conventional T cells in the presence of excessive co-stimulation when murine T cells are stimulated in vitro with allogeneic antigen-presenting cells (APCs). Antigen-specific Tregs increase the number of cells interacting with dendritic cells (DCs) by increasing the number of viable DCs and the expression of adhesion molecules on DCs. Theoretical simulations and mathematical models representing the dynamics of T-APC interaction and T-cell numbers in a lymph node indicate that Tregs reduce the dissociation probability of T cells from APCs and increase the new association. These functions contribute to tolerance by enhancing the interaction of low-affinity T cells with APCs. Supporting the theoretical analyses, we found that reducing the T-cell numbers in mice increases the ratio of specific T cells among CD4+ T cells after immunization and effectively induces autoimmune diabetes in non obese diabetes mice. Thus, as a critical function, antigen-specific Tregs stabilize the immune state, irrespective of it being tolerant or responsive, by augmenting T-APC interaction. We propose a novel regulation model in which stable tolerance with large heterogeneous populations proceeds to a specific immune response through a transient state with few populations.
Collapse
Affiliation(s)
- Tomoyuki Yamaguchi
- Basic Immunology Laboratory, Research Institute, Nozaki Tokushukai Hospital, Tanigawa, Daito, Osaka, Japan
- Single Molecule Imaging, WPI Immunology Frontier Research Center, Osaka University, Furuedai, Suita, Osaka, Japan
- Quantitative Biology Center, RIKEN, Furuedai, Suita, Osaka, Japan
| | - Shunsuke Teraguchi
- Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | - Chikara Furusawa
- Quantitative Biology Center, RIKEN, Furuedai, Suita, Osaka, Japan
- Universal Biology Institute, University of Tokyo, Hongo, Tokyo, Japan
| | - Hiroaki Machiyama
- Single Molecule Imaging, WPI Immunology Frontier Research Center, Osaka University, Furuedai, Suita, Osaka, Japan
- Quantitative Biology Center, RIKEN, Furuedai, Suita, Osaka, Japan
| | | | - Hideaki Fujita
- Single Molecule Imaging, WPI Immunology Frontier Research Center, Osaka University, Furuedai, Suita, Osaka, Japan
- Quantitative Biology Center, RIKEN, Furuedai, Suita, Osaka, Japan
| | - Shimon Sakaguchi
- Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | - Toshio Yanagida
- Single Molecule Imaging, WPI Immunology Frontier Research Center, Osaka University, Furuedai, Suita, Osaka, Japan
- Quantitative Biology Center, RIKEN, Furuedai, Suita, Osaka, Japan
| |
Collapse
|
36
|
Abstract
The repertoire of αβ T cell antigen receptors (TCRs) on mature T cells is selected in the thymus where it is rendered both self-tolerant and restricted to the recognition of major histocompatibility complex molecules presenting peptide antigens (pMHC). It remains unclear whether germline TCR sequences exhibit an inherent bias to interact with pMHC prior to selection. Here, we isolated TCR libraries from unselected thymocytes and upon reexpression of these random TCR repertoires in recipient T cell hybridomas, interrogated their reactivities to antigen-presenting cell lines. While these random TCR combinations could potentially have reacted with any surface molecule on the cell lines, the hybridomas were stimulated most frequently by pMHC ligands. The nature and CDR3 loop composition of the TCRβ chain played a dominant role in determining pMHC-reactivity. Replacing the germline regions of mouse TCRβ chains with those of other jawed vertebrates preserved reactivity to mouse pMHC. Finally, introducing the CD4 coreceptor into the hybridomas increased the proportion of cells that could respond to pMHC ligands. Thus, αβ TCRs display an intrinsic and evolutionary conserved bias for pMHC molecules in the absence of any selective pressure, which is further strengthened in the presence of coreceptors.
Collapse
|
37
|
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
|
38
|
Oh-Hora M, Lu X, Shiokawa M, Takayanagi H, Yamasaki S. Stromal Interaction Molecule Deficiency in T Cells Promotes Spontaneous Follicular Helper T Cell Development and Causes Type 2 Immune Disorders. THE JOURNAL OF IMMUNOLOGY 2019; 202:2616-2627. [PMID: 30910863 DOI: 10.4049/jimmunol.1700610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 03/03/2019] [Indexed: 12/24/2022]
Abstract
Appropriate T cell responses are controlled by strict balance between activatory and inhibitory pathways downstream of TCR. Although mice or humans with impaired TCR signaling develop autoimmunity, the precise molecular mechanisms linking reduced TCR signaling to autoimmunity are not fully understood. Engagement of TCR activates Ca2+ signaling mainly through store-operated Ca2+ entry activated by stromal interaction molecule (Stim) 1 and Stim2. Despite defective T cell activation, mice deficient in both Stim1 and Stim2 in T cells (conditional double knockout [cDKO]) developed lymphoproliferative disorders and skin inflammation with a concomitant increase in serum IgG1 and IgE levels. In cDKO mice, follicular helper T (Tfh) cells were dramatically increased in number, and they produced IL-4 spontaneously. These inflammatory symptoms were abolished by the deletion of IL-4 in cDKO mice. Tfh development and inflammatory symptoms in cDKO mice were abrogated by further deletion of NFAT2 in T cells. These findings suggest that Tfh cells spontaneously developed in the absence of Ca2+ signaling and caused unregulated type 2 responses.
Collapse
Affiliation(s)
- Masatsugu Oh-Hora
- Division of Molecular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; .,Department of Biochemistry, Juntendo University School of Medicine, Tokyo 113-8421, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Xiuyuan Lu
- Division of Molecular and Cellular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Moe Shiokawa
- Division of Molecular and Cellular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; and
| | - Sho Yamasaki
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan; .,Division of Molecular and Cellular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan.,Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| |
Collapse
|
39
|
Christopoulos P, Chung I, Bozorgmehr F, Muley T, Meister M, Kobinger S, Marx A, Thomas M, Winter H, Herpel E, Rieker RJ, Fisch P, Grosch H. Deficient CD247 expression is a typical histopathological characteristic of thymomas with cortical features. Histopathology 2018; 73:1040-1043. [PMID: 30079467 DOI: 10.1111/his.13724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Inn Chung
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Farastuk Bozorgmehr
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Meister
- Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Sonja Kobinger
- Department of Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Marx
- Institute of Pathology, Mannheim University Hospital, Mannheim, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Ralf J Rieker
- Institute of Pathology, University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Paul Fisch
- Department of Pathology, Freiburg University Hospital, Freiburg, Germany
| | - Heidrun Grosch
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
40
|
James JR. Tuning ITAM multiplicity on T cell receptors can control potency and selectivity to ligand density. Sci Signal 2018; 11:11/531/eaan1088. [PMID: 29789296 DOI: 10.1126/scisignal.aan1088] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The T cell antigen receptor (TCR) recognizes peptides from pathogenic proteins bound in the major histocompatibility complex (MHC). To convert this binding event into downstream signaling, the TCR complex contains immunoreceptor tyrosine-based activation motifs (ITAMs) that act as docking sites for the cytoplasmic tyrosine kinase ZAP-70. Unique among antigen receptors, the TCR complex uses 10 ITAMs to transduce peptide-MHC binding to the cell interior. Using synthetic, drug-inducible receptor-ligand pairs, it was found that greater ITAM multiplicity primarily enhanced the efficiency with which ligand binding was converted into an intracellular signal. This manifested as an increase in the fraction of cells that became activated in response to antigen, and a more synchronous initiation of TCR-proximal signaling, rather than direct amplification of the intracellular signals. Exploiting these findings, the potency and selectivity of chimeric antigen receptors targeted against cancer were substantially enhanced by modulating the number of encoded ITAMs.
Collapse
Affiliation(s)
- John R James
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 OQH, UK.
| |
Collapse
|
41
|
|
42
|
Glassman CR, Parrish HL, Lee MS, Kuhns MS. Reciprocal TCR-CD3 and CD4 Engagement of a Nucleating pMHCII Stabilizes a Functional Receptor Macrocomplex. Cell Rep 2018; 22:1263-1275. [PMID: 29386113 PMCID: PMC5813697 DOI: 10.1016/j.celrep.2017.12.104] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/07/2017] [Accepted: 12/27/2017] [Indexed: 12/24/2022] Open
Abstract
CD4+ T cells convert the time that T cell receptors (TCRs) interact with peptides embedded within class II major histocompatibility complex molecules (pMHCII) into signals that direct cell-fate decisions. In principle, TCRs relay information to intracellular signaling motifs of the associated CD3 subunits, while CD4 recruits the kinase Lck to those motifs upon coincident detection of pMHCII. But the mechanics by which this occurs remain enigmatic. In one model, the TCR and CD4 bind pMHCII independently, while in another, CD4 interacts with a composite surface formed by the TCR-CD3 complex bound to pMHCII. Here, we report that the duration of TCR-pMHCII interactions impact CD4 binding to MHCII. In turn, CD4 increases TCR confinement to pMHCII via reciprocal interactions involving membrane distal and proximal CD4 ectodomains. The data suggest that a precisely assembled macrocomplex functions to reliably convert TCR-pMHCII confinement into reproducible signals that orchestrate adaptive immunity.
Collapse
Affiliation(s)
- Caleb R Glassman
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Heather L Parrish
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Mark S Lee
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Michael S Kuhns
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA; The BIO-5 Institute, The University of Arizona College of Medicine, Tucson, AZ 85724, USA; The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, AZ 85724, USA; The University of Arizona Cancer Center, The University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| |
Collapse
|
43
|
An optimized single chain TCR scaffold relying on the assembly with the native CD3-complex prevents residual mispairing with endogenous TCRs in human T-cells. Oncotarget 2018; 7:21199-221. [PMID: 27028870 PMCID: PMC5008279 DOI: 10.18632/oncotarget.8385] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/10/2016] [Indexed: 12/29/2022] Open
Abstract
Immunotherapy of cancer envisions the adoptive transfer of T-cells genetically engineered with tumor-specific heterodimeric α/β T-cell receptors (TCRα/β). However, potential mispairing of introduced TCRα/β-chains with endogenous β/α-ones may evoke unpredictable autoimmune reactivities. A novel single chain (sc)TCR format relies on the fusion of the Vα-Linker-Vβ-fragment to the TCR Cβ-domain and coexpression of the TCR Cα-domain capable of recruiting the natural CD3-complex for full and hence, native T-cell signaling. Here, we tested whether such a gp100(280-288)- or p53(264-272) tumor antigen-specific scTCR is still prone to mispairing with TCRα. In a human Jurkat-76 T-cell line lacking endogenous TCRs, surface expression and function of a scTCR could be reconstituted by any cointroduced TCRα-chain indicating mispairing to take place on a molecular basis. In contrast, transduction into human TCRα/β-positive T-cells revealed that mispairing is largely reduced. Competition experiments in Jurkat-76 confirmed the preference of dcTCR to selfpair and to spare scTCR. This also allowed for the generation of dc/scTCR-modified cytomegalovirus/tumor antigen-bispecific T-cells to augment T-cell activation in CMV-infected tumor patients. Residual mispairing was prevented by strenghtening the Vα-Li-Vβ-fragment through the design of a novel disulfide bond between a Vα- and a linker-resident residue close to Vβ. Multimer-stainings, and cytotoxicity-, IFNγ-secretion-, and CFSE-proliferation-assays, the latter towards dendritic cells endogenously processing RNA-electroporated gp100 antigen proved the absence of hybrid scTCR/TCRα-formation without impairing avidity of scTCR/Cα in T-cells. Moreover, a fragile cytomegalovirus pp65(495-503)-specific scTCR modified this way acquired enhanced cytotoxicity. Thus, optimized scTCR/Cα inhibits residual TCR mispairing to accomplish safe adoptive immunotherapy for bulk endogenous TCRα/β-positive T-cells.
Collapse
|
44
|
Ngoenkam J, Schamel WW, Pongcharoen S. Selected signalling proteins recruited to the T-cell receptor-CD3 complex. Immunology 2018; 153:42-50. [PMID: 28771705 PMCID: PMC5721247 DOI: 10.1111/imm.12809] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/26/2017] [Accepted: 07/27/2017] [Indexed: 12/22/2022] Open
Abstract
The T-cell receptor (TCR)-CD3 complex, expressed on T cells, determines the outcome of a T-cell response. It consists of the TCR-αβ heterodimer and the non-covalently associated signalling dimers of CD3εγ, CD3εδ and CD3ζζ. TCR-αβ binds specifically to a cognate peptide antigen bound to an MHC molecule, whereas the CD3 subunits transmit the signal into the cytosol to activate signalling events. Recruitment of proteins to specialized localizations is one mechanism to regulate activation and termination of signalling. In the last 25 years a large number of signalling molecules recruited to the TCR-CD3 complex upon antigen binding to TCR-αβ have been described. Here, we review knowledge about five of those interaction partners: Lck, ZAP-70, Nck, WASP and Numb. Some of these proteins have been targeted in the development of immunomodulatory drugs aiming to treat patients with autoimmune diseases and organ transplants.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- CD3 Complex/chemistry
- CD3 Complex/genetics
- CD3 Complex/metabolism
- Carrier Proteins/chemistry
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Humans
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism
- Membrane Proteins/metabolism
- Mutation
- Nerve Tissue Proteins/metabolism
- Oncogene Proteins/metabolism
- Protein Binding
- Protein Interaction Domains and Motifs
- Receptor-CD3 Complex, Antigen, T-Cell/chemistry
- Receptor-CD3 Complex, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Wiskott-Aldrich Syndrome Protein/metabolism
- ZAP-70 Protein-Tyrosine Kinase/metabolism
Collapse
Affiliation(s)
- Jatuporn Ngoenkam
- Department of Microbiology and ParasitologyFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| | - Wolfgang W. Schamel
- Department of ImmunologyInstitute for Biology IIIFaculty of BiologyUniversity of FreiburgFreiburgGermany
- BIOSS Centre for Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
- Centre for Chronic Immunodeficiency (CCI)Medical Centre‐University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Sutatip Pongcharoen
- Centre of Excellence in Medical BiotechnologyFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
- Centre of Excellence in Petroleum, Petrochemicals and Advanced MaterialsFaculty of ScienceNaresuan UniversityPhitsanulokThailand
- Department of MedicineFaculty of MedicineNaresuan UniversityPhitsanulokThailand
| |
Collapse
|
45
|
Bettini ML, Chou PC, Guy CS, Lee T, Vignali KM, Vignali DAA. Cutting Edge: CD3 ITAM Diversity Is Required for Optimal TCR Signaling and Thymocyte Development. THE JOURNAL OF IMMUNOLOGY 2017; 199:1555-1560. [PMID: 28733484 DOI: 10.4049/jimmunol.1700069] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/27/2017] [Indexed: 12/21/2022]
Abstract
For the αβ or γδTCR chains to integrate extracellular stimuli into the appropriate intracellular cellular response, they must use the 10 ITAMs found within the CD3 subunits (CD3γε, CD3δε, and ζζ) of the TCR signaling complex. However, it remains unclear whether each specific ITAM sequence of the individual subunit (γεδζ) is required for thymocyte development or whether any particular CD3 ITAM motif is sufficient. In this article, we show that mice utilizing a single ITAM sequence (γ, ε, δ, ζa, ζb, or ζc) at each of the 10 ITAM locations exhibit a substantial reduction in thymic cellularity and limited CD4-CD8- (double-negative) to CD4+CD8+ (double-positive) maturation because of low TCR expression and signaling. Together, the data suggest that ITAM sequence diversity is required for optimal TCR signal transduction and subsequent T cell maturation.
Collapse
Affiliation(s)
- Matthew L Bettini
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105; .,Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030
| | - Po-Chein Chou
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Clifford S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Thomas Lee
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030
| | - Kate M Vignali
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; and
| | - Dario A A Vignali
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105; .,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; and.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| |
Collapse
|
46
|
Mukhopadhyay H, de Wet B, Clemens L, Maini PK, Allard J, van der Merwe PA, Dushek O. Multisite Phosphorylation Modulates the T Cell Receptor ζ-Chain Potency but not the Switchlike Response. Biophys J 2017; 110:1896-1906. [PMID: 27119648 PMCID: PMC4850346 DOI: 10.1016/j.bpj.2016.03.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/08/2016] [Accepted: 03/15/2016] [Indexed: 11/17/2022] Open
Abstract
Multisite phosphorylation is ubiquitous in cellular signaling and is thought to provide signaling proteins with additional regulatory mechanisms. Indeed, mathematical models have revealed a large number of mechanisms by which multisite phosphorylation can produce switchlike responses. The T cell antigen receptor (TCR) is a multisubunit receptor on the surface of T cells that is a prototypical multisite substrate as it contains 20 sites that are distributed on 10 conserved immunoreceptor tyrosine-based activation motifs (ITAMs). The TCR ζ-chain is a homodimer subunit that contains six ITAMs (12 sites) and exhibits a number of properties that are predicted to be sufficient for a switchlike response. We have used cellular reconstitution to systematically study multisite phosphorylation of the TCR ζ-chain. We find that multisite phosphorylation proceeds by a nonsequential random mechanism, and find no evidence that multiple ITAMs modulate a switchlike response but do find that they alter receptor potency and maximum phosphorylation. Modulation of receptor potency can be explained by a reduction in molecular entropy of the disordered ζ-chain upon phosphorylation. We further find that the tyrosine kinase ZAP-70 increases receptor potency but does not modulate the switchlike response. In contrast to other multisite proteins, where phosphorylations act in strong concert to modulate protein function, we suggest that the multiple ITAMs on the TCR function mainly to amplify subsequent signaling.
Collapse
Affiliation(s)
- Himadri Mukhopadhyay
- Sir William Dunn School of Pathology, Mathematical Institute, University of Oxford, Oxfordshire, United Kingdom
| | - Ben de Wet
- Sir William Dunn School of Pathology, Mathematical Institute, University of Oxford, Oxfordshire, United Kingdom
| | - Lara Clemens
- Department of Mathematics, University of California-Irvine, Irvine, California
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxfordshire, United Kingdom
| | - Jun Allard
- Department of Mathematics, University of California-Irvine, Irvine, California
| | - P Anton van der Merwe
- Sir William Dunn School of Pathology, Mathematical Institute, University of Oxford, Oxfordshire, United Kingdom.
| | - Omer Dushek
- Sir William Dunn School of Pathology, Mathematical Institute, University of Oxford, Oxfordshire, United Kingdom; Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxfordshire, United Kingdom.
| |
Collapse
|
47
|
Holmberg D, Ruikka K, Lindgren P, Eliasson M, Mayans S. Association of CD247 (CD3ζ) gene polymorphisms with T1D and AITD in the population of northern Sweden. BMC MEDICAL GENETICS 2016; 17:70. [PMID: 27716086 PMCID: PMC5050583 DOI: 10.1186/s12881-016-0333-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/29/2016] [Indexed: 12/31/2022]
Abstract
Background T1D and AITD are autoimmune disorders commonly occurring in the same family and even in the same individual. The genetic contribution to these disorders is complex making uncovering of susceptibility genes very challenging. The general aim of this study was to identify loci and genes contributing to T1D/AITD susceptibility. Our strategy was to perform linkage and association studies in the relatively genetically homogenous population of northern Sweden. We performed a GWLS to find genomic regions linked to T1D/AITD in families from northern Sweden and we performed an association study in the families to test for association between T1D/AITD and variants in previously published candidate genes as well as a novel candidate gene, CD247. Methods DNA prepared from 459 individuals was used to perform a linkage and an association study. The ABI PRISM Linkage Mapping Set v2.5MD10 was employed for an initial 10-cM GWLS, and additional markers were added for fine mapping. Merlin was used for linkage calculations. For the association analysis, a GoldenGate Custom Panel from Illumina containing 79 SNPs of interest was used and FBAT was used for association calculations. Results Our study revealed linkage to two previously identified chromosomal regions, 4q25 and 6p22, as well as to a novel chromosomal region, 1q23. The association study replicated association to PTPN22, HLA-DRB1, INS, IFIH1, CTLA4 and C12orf30. Evidence in favor of association was also found for SNPs in the novel susceptibility gene CD247. Conclusions Several risk loci for T1D/AITD identified in published association studies were replicated in a family material, of modest size, from northern Sweden. This provides evidence that these loci confer disease susceptibility in this population and emphasizes that small to intermediate sized family studies in this population can be used in a cost-effective manner for the search of genes involved in complex diseases. The linkage study revealed a chromosomal region in which a novel T1D/AITD susceptibility gene, CD247, is located. The association study showed association between T1D/AITD and several variants in this gene. These results suggests that common susceptibility genes act in concert with variants of CD247 to generate genetic risk for T1D/AITD in this population. Electronic supplementary material The online version of this article (doi:10.1186/s12881-016-0333-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dan Holmberg
- Department of Medical Biosciences - Medical Genetics, Umeå University, SE-901 85, Umeå, Sweden.,EMV, Immunology, BMC, Lund University, SE-221 00, Lund, Sweden
| | - Karin Ruikka
- Department of Medicine, Sunderby Hospital, SE-971 80, Luleå, Sweden
| | - Petter Lindgren
- Department of Medical Biosciences - Medical Genetics, Umeå University, SE-901 85, Umeå, Sweden
| | - Mats Eliasson
- Department of Medicine, Sunderby Hospital, SE-971 80, Luleå, Sweden.,Department of Public Health and Clinical Medicine, Umeå University, SE-901 85, Umeå, Sweden
| | - Sofia Mayans
- Department of Medical Biosciences - Medical Genetics, Umeå University, SE-901 85, Umeå, Sweden. .,Department of Clinical Microbiology, Division of Immunology, Umeå University, Building 6C, SE-90185, Umeå, Sweden.
| |
Collapse
|
48
|
Functional role of T-cell receptor nanoclusters in signal initiation and antigen discrimination. Proc Natl Acad Sci U S A 2016; 113:E5454-63. [PMID: 27573839 DOI: 10.1073/pnas.1607436113] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Antigen recognition by the T-cell receptor (TCR) is a hallmark of the adaptive immune system. When the TCR engages a peptide bound to the restricting major histocompatibility complex molecule (pMHC), it transmits a signal via the associated CD3 complex. How the extracellular antigen recognition event leads to intracellular phosphorylation remains unclear. Here, we used single-molecule localization microscopy to quantify the organization of TCR-CD3 complexes into nanoscale clusters and to distinguish between triggered and nontriggered TCR-CD3 complexes. We found that only TCR-CD3 complexes in dense clusters were phosphorylated and associated with downstream signaling proteins, demonstrating that the molecular density within clusters dictates signal initiation. Moreover, both pMHC dose and TCR-pMHC affinity determined the density of TCR-CD3 clusters, which scaled with overall phosphorylation levels. Thus, TCR-CD3 clustering translates antigen recognition by the TCR into signal initiation by the CD3 complex, and the formation of dense signaling-competent clusters is a process of antigen discrimination.
Collapse
|
49
|
Lee T, Shevchenko I, Sprouse ML, Bettini M, Bettini ML. Retroviral Transduction of Bone Marrow Progenitor Cells to Generate T-cell Receptor Retrogenic Mice. J Vis Exp 2016. [PMID: 27500835 DOI: 10.3791/54196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
T cell receptor (TCR) signaling is essential in the development and differentiation of T cells in the thymus and periphery, respectively. The vast array of TCRs proves studying a specific antigenic response difficult. Therefore, TCR transgenic mice were made to study positive and negative selection in the thymus as well as peripheral T cell activation, proliferation and tolerance. However, relatively few TCR transgenic mice have been generated specific to any given antigen. Thus, studies involving TCRs of varying affinities for the same antigenic peptide have been lacking. The generation of a new TCR transgenic line can take six or more months. Additionally, any specific backcrosses can take an additional six months. In order to allow faster generation and screening of multiple TCRs, a protocol for retroviral transduction of bone marrow was established with stoichiometric expression of the TCRα and TCRβ chains and the generation of retrogenic mice. Each retrogenic mouse is essentially a founder, virtually negating a founder effect, while the length of time to generate a TCR retrogenic is cut from six months to approximately six weeks. Here we present a rapid and flexible alternative to TCR transgenic mice that can be expressed on any chosen background with any particular TCR.
Collapse
Affiliation(s)
- Thomas Lee
- Department of Pediatrics, Baylor College of Medicine
| | | | | | - Maria Bettini
- Department of Pediatrics, Baylor College of Medicine
| | | |
Collapse
|
50
|
Corrado M, Mariotti FR, Trapani L, Taraborrelli L, Nazio F, Cianfanelli V, Soriano ME, Schrepfer E, Cecconi F, Scorrano L, Campello S. Macroautophagy inhibition maintains fragmented mitochondria to foster T cell receptor-dependent apoptosis. EMBO J 2016; 35:1793-809. [PMID: 27390127 DOI: 10.15252/embj.201593727] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/31/2016] [Indexed: 01/08/2023] Open
Abstract
Mitochondrial dynamics and functionality are linked to the autophagic degradative pathway under several stress conditions. However, the interplay between mitochondria and autophagy upon cell death signalling remains unclear. The T-cell receptor pathway signals the so-called activation-induced cell death (AICD) essential for immune tolerance regulation. Here, we show that this apoptotic pathway requires the inhibition of macroautophagy. Protein kinase-A activation downstream of T-cell receptor signalling inhibits macroautophagy upon AICD induction. This leads to the accumulation of damaged mitochondria, which are fragmented, display remodelled cristae and release cytochrome c, thereby driving apoptosis. Autophagy-forced reactivation that clears the Parkin-decorated mitochondria is as effective in inhibiting apoptosis as genetic interference with cristae remodelling and cytochrome c release. Thus, upon AICD induction regulation of macroautophagy, rather than selective mitophagy, ensures apoptotic progression.
Collapse
Affiliation(s)
- Mauro Corrado
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Padova, Italy IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | | | - Lucia Taraborrelli
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Francesca Nazio
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Valentina Cianfanelli
- Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Emilie Schrepfer
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Padova, Italy Department of Biology, University of Padova, Padova, Italy
| | - Francesco Cecconi
- IRCCS Fondazione Santa Lucia, Rome, Italy Department of Biology, University of Rome Tor Vergata, Rome, Italy Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Luca Scorrano
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Padova, Italy Department of Biology, University of Padova, Padova, Italy
| | - Silvia Campello
- IRCCS Fondazione Santa Lucia, Rome, Italy Department of Biology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|