Molecular Mechanisms of Early T Cell Signaling

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cell Signaling".

Deadline for manuscript submissions: closed (28 February 2021) | Viewed by 25261

Special Issue Editors


E-Mail Website
Guest Editor
Institute of Applied Physics, TU Wien, Getreidemarkt 9, A-1060 Wien, Austria
Interests: single molecule microscopy; super-resolution microscopy; membrane biophysics; diffusion; T cell signaling

E-Mail Website
Guest Editor
Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, Lazarettgasse 19, A-1990 Vienna, Austria
Interests: T-cell antigen recognition; advanced molecular imaging; synthetic biology; systems biology

Special Issue Information

Dear Colleagues,

A remarkable feature of our immune system is its inherent ability to distinguish harmful from harmless based on the primary protein structure of antigens. T-cells embody this trait of adaptive immunity through their unique detection of peptide antigens, driven by αβT-cell receptors (TCRs) on the T-cell binding to particular peptide antigen-loaded MHC molecules (pMHC) displayed by antigen-presenting cells. One of the most fascinating aspects of antigen recognition is its exquisite sensitivity combined with enormous efficacy: T cells can detect even a single antigenic pMHC molecule among a great number of structurally similar yet non-stimulatory pMHCs. The molecular/cellular mechanisms underlying this remarkable quality, however, are not at all understood, even though their relevance in both disease progression and intervention can hardly be overestimated: Inappropriate changes in T-cell reactivity can be harmful if not fatal, as they can weaken the body’s defense against pathogens and cancer and cause allergies or trigger autoimmunity.

In recent years, results from novel experimental approaches have stimulated the formulation of new concepts explaining antigen recognition and brought new parameters to our attention, such as protein size exclusion effects, spatiotemporal organization of molecules, tensile forces acting between TCR and pMHC, and cooperative mechanisms. This Special Issue shall provide an overview of the state-of-the-art, ranging from method development to model conceptualization and synthetic approaches to capitalize on these features for immune intervention in a clinical setting.

Prof. Dr. Gerhard J. Schütz
Prof. Dr. Johannes B. Huppa
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • T cell signaling
  • Immunological synapse
  • TCR–CD3 complex
  • MHC

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

12 pages, 2896 KiB  
Article
Cooperative Stabilization of Close-Contact Zones Leads to Sensitivity and Selectivity in T-Cell Recognition
by Bartosz Różycki and Thomas R. Weikl
Cells 2021, 10(5), 1023; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10051023 - 26 Apr 2021
Cited by 4 | Viewed by 2006
Abstract
T cells are sensitive to 1 to 10 foreign-peptide-MHC complexes among a vast majority of self-peptide-MHC complexes, and discriminate selectively between peptide-MHC complexes that differ not much in their binding affinity to T-cell receptors (TCRs). Quantitative models that aim to explain this sensitivity [...] Read more.
T cells are sensitive to 1 to 10 foreign-peptide-MHC complexes among a vast majority of self-peptide-MHC complexes, and discriminate selectively between peptide-MHC complexes that differ not much in their binding affinity to T-cell receptors (TCRs). Quantitative models that aim to explain this sensitivity and selectivity largely focus on single TCR/peptide-MHC complexes, but T cell adhesion involves a multitude of different complexes. In this article, we demonstrate in a three-dimensional computational model of T-cell adhesion that the cooperative stabilization of close-contact zones is sensitive to one to three foreign-peptide-MHC complexes and occurs at a rather sharp threshold affinity of these complexes, which implies selectivity. In these close-contact zones with lateral extensions of hundred to several hundred nanometers, few TCR/foreign-peptide-MHC complexes and many TCR/self-peptide-MHC complexes are segregated from LFA-1/ICAM-1 complexes that form at larger membrane separations. Previous high-resolution microscopy experiments indicate that the sensitivity and selectivity in the formation of closed-contact zones reported here are relevant for T-cell recognition, because the stabilization of close-contact zones by foreign, agonist peptide-MHC complexes precedes T-cell signaling and activation in the experiments. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Early T Cell Signaling)
Show Figures

Figure 1

13 pages, 1459 KiB  
Article
Cooperative Interaction of Nck and Lck Orchestrates Optimal TCR Signaling
by Frederike A. Hartl, Jatuporn Ngoenkam, Esmeralda Beck-Garcia, Liz Cerqueira, Piyamaporn Wipa, Pussadee Paensuwan, Prapat Suriyaphol, Pankaj Mishra, Burkhart Schraven, Stefan Günther, Sutatip Pongcharoen, Wolfgang W. A. Schamel and Susana Minguet
Cells 2021, 10(4), 834; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10040834 - 7 Apr 2021
Cited by 5 | Viewed by 3966
Abstract
The T cell antigen receptor (TCR) is expressed on T cells, which orchestrate adaptive immune responses. It is composed of the ligand-binding clonotypic TCRαβ heterodimer and the non-covalently bound invariant signal-transducing CD3 complex. Among the CD3 subunits, the CD3ε cytoplasmic tail contains binding [...] Read more.
The T cell antigen receptor (TCR) is expressed on T cells, which orchestrate adaptive immune responses. It is composed of the ligand-binding clonotypic TCRαβ heterodimer and the non-covalently bound invariant signal-transducing CD3 complex. Among the CD3 subunits, the CD3ε cytoplasmic tail contains binding motifs for the Src family kinase, Lck, and the adaptor protein, Nck. Lck binds to a receptor kinase (RK) motif and Nck binds to a proline-rich sequence (PRS). Both motifs only become accessible upon ligand binding to the TCR and facilitate the recruitment of Lck and Nck independently of phosphorylation of the TCR. Mutations in each of these motifs cause defects in TCR signaling and T cell activation. Here, we investigated the role of Nck in proximal TCR signaling by silencing both Nck isoforms, Nck1 and Nck2. In the absence of Nck, TCR phosphorylation, ZAP70 recruitment, and ZAP70 phosphorylation was impaired. Mechanistically, this is explained by loss of Lck recruitment to the stimulated TCR in cells lacking Nck. Hence, our data uncover a previously unknown cooperative interaction between Lck and Nck to promote optimal TCR signaling. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Early T Cell Signaling)
Show Figures

Figure 1

16 pages, 2576 KiB  
Article
CD8 Co-Receptor Enhances T-Cell Activation without Any Effect on Initial Attachment
by Philippe Robert, Laurent Limozin, P. Anton van der Merwe and Pierre Bongrand
Cells 2021, 10(2), 429; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10020429 - 18 Feb 2021
Cited by 1 | Viewed by 1993
Abstract
The scanning of surrounding tissues by T lymphocytes to detect cognate antigens requires high speed, sensitivity and specificity. T-cell receptor (TCR) co-receptors such as CD8 increase detection performance, but the exact mechanism remains incompletely understood. Here, we used a laminar flow chamber to [...] Read more.
The scanning of surrounding tissues by T lymphocytes to detect cognate antigens requires high speed, sensitivity and specificity. T-cell receptor (TCR) co-receptors such as CD8 increase detection performance, but the exact mechanism remains incompletely understood. Here, we used a laminar flow chamber to measure at the single molecule level the kinetics of bond formation and rupture between TCR- transfected CD8+ and CD8− Jurkat cells and surfaces coated with five peptide-exposing major histocompatibility antigens (pMHCs) of varying activating power. We also used interference reflection microscopy to image the spreading of these cells dropped on pMHC-exposing surfaces. CD8 did not influence the TCR–pMHC interaction during the first few seconds following cell surface encounter, but it promoted the subsequent spreading responses, suggesting that CD8 was involved in early activation rather than binding. Further, the rate and extent of spreading, but not the lag between contact and spreading initiation, depended on the pMHC. Elucidating T-lymphocyte detection strategy may help unravel underlying signaling networks. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Early T Cell Signaling)
Show Figures

Figure 1

24 pages, 14085 KiB  
Article
A LAT-Based Signaling Complex in the Immunological Synapse as Determined with Live Cell Imaging Is Less Stable in T Cells with Regulatory Capability
by Yikui Li, Helen M. Tunbridge, Graham J. Britton, Elaine V. Hill, Parisa Sinai, Silvia Cirillo, Clare Thompson, Farnaz Fallah-Arani, Simon J. Dovedi, David C. Wraith and Christoph Wülfing
Cells 2021, 10(2), 418; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10020418 - 17 Feb 2021
Cited by 2 | Viewed by 2485
Abstract
Peripheral immune regulation is critical for the maintenance of self-tolerance. Here we have investigated signaling processes that distinguish T cells with regulatory capability from effector T cells. The murine Tg4 T cell receptor recognizes a peptide derived from the self-antigen myelin basic protein. [...] Read more.
Peripheral immune regulation is critical for the maintenance of self-tolerance. Here we have investigated signaling processes that distinguish T cells with regulatory capability from effector T cells. The murine Tg4 T cell receptor recognizes a peptide derived from the self-antigen myelin basic protein. T cells from Tg4 T cell receptor transgenic mice can be used to generate effector T cells and three types of T cells with regulatory capability, inducible regulatory T cells, T cells tolerized by repeated in vivo antigenic peptide exposure or T cells treated with the tolerogenic drug UCB9608 (a phosphatidylinositol 4 kinase IIIβ inhibitor). We comparatively studied signaling in all of these T cells by activating them with the same antigen presenting cells presenting the same myelin basic protein peptide. Supramolecular signaling structures, as efficiently detected by large-scale live cell imaging, are critical mediators of T cell activation. The formation of a supramolecular signaling complex anchored by the adaptor protein linker for activation of T cells (LAT) was consistently terminated more rapidly in Tg4 T cells with regulatory capability. Such termination could be partially reversed by blocking the inhibitory receptors CTLA-4 and PD-1. Our work suggests that attenuation of proximal signaling may favor regulatory over effector function in T cells. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Early T Cell Signaling)
Show Figures

Graphical abstract

18 pages, 3472 KiB  
Article
Retrograde and Anterograde Transport of Lat-Vesicles during the Immunological Synapse Formation: Defining the Finely-Tuned Mechanism
by Juan José Saez, Stephanie Dogniaux, Massiullah Shafaq-Zadah, Ludger Johannes, Claire Hivroz and Andrés Ernesto Zucchetti
Cells 2021, 10(2), 359; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10020359 - 9 Feb 2021
Cited by 4 | Viewed by 3471
Abstract
LAT is an important player of the signaling cascade induced by TCR activation. This adapter molecule is present at the plasma membrane of T lymphocytes and more abundantly in intracellular compartments. Upon T cell activation the intracellular pool of LAT is recruited to [...] Read more.
LAT is an important player of the signaling cascade induced by TCR activation. This adapter molecule is present at the plasma membrane of T lymphocytes and more abundantly in intracellular compartments. Upon T cell activation the intracellular pool of LAT is recruited to the immune synapse (IS). We previously described two pathways controlling LAT trafficking: retrograde transport from endosomes to the TGN, and anterograde traffic from the Golgi to the IS. We address the specific role of four proteins, the GTPase Rab6, the t-SNARE syntaxin-16, the v-SNARE VAMP7 and the golgin GMAP210, in each pathway. Using different methods (endocytosis and Golgi trap assays, confocal and TIRF microscopy, TCR-signalosome pull down) we show that syntaxin-16 is regulating the retrograde transport of LAT whereas VAMP7 is regulating the anterograde transport. Moreover, GMAP210 and Rab6, known to contribute to both pathways, are in our cellular context, specifically and respectively, involved in anterograde and retrograde transport of LAT. Altogether, our data describe how retrograde and anterograde pathways coordinate LAT enrichment at the IS and point to the Golgi as a central hub for the polarized recruitment of LAT to the IS. The role that this finely-tuned transport of signaling molecules plays in T-cell activation is discussed. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Early T Cell Signaling)
Show Figures

Figure 1

20 pages, 2367 KiB  
Article
A Novel, LAT/Lck Double Deficient T Cell Subline J.CaM1.7 for Combined Analysis of Early TCR Signaling
by Inmaculada Vico-Barranco, Mikel M. Arbulo-Echevarria, Isabel Serrano-García, Alba Pérez-Linaza, José M. Miranda-Sayago, Arkadiusz Miazek, Isaac Narbona-Sánchez and Enrique Aguado
Cells 2021, 10(2), 343; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10020343 - 6 Feb 2021
Cited by 4 | Viewed by 3055
Abstract
Intracellular signaling through the T cell receptor (TCR) is essential for T cell development and function. Proper TCR signaling requires the sequential activities of Lck and ZAP-70 kinases, which result in the phosphorylation of tyrosine residues located in the CD3 ITAMs and the [...] Read more.
Intracellular signaling through the T cell receptor (TCR) is essential for T cell development and function. Proper TCR signaling requires the sequential activities of Lck and ZAP-70 kinases, which result in the phosphorylation of tyrosine residues located in the CD3 ITAMs and the LAT adaptor, respectively. LAT, linker for the activation of T cells, is a transmembrane adaptor protein that acts as a scaffold coupling the early signals coming from the TCR with downstream signaling pathways leading to cellular responses. The leukemic T cell line Jurkat and its derivative mutants J.CaM1.6 (Lck deficient) and J.CaM2 (LAT deficient) have been widely used to study the first signaling events upon TCR triggering. In this work, we describe the loss of LAT adaptor expression found in a subline of J.CaM1.6 cells and analyze cis-elements responsible for the LAT expression defect. This new cell subline, which we have called J.CaM1.7, can re-express LAT adaptor after Protein Kinase C (PKC) activation, which suggests that activation-induced LAT expression is not affected in this new cell subline. Contrary to J.CaM1.6 cells, re-expression of Lck in J.CaM1.7 cells was not sufficient to recover TCR-associated signals, and both LAT and Lck had to be introduced to recover activatory intracellular signals triggered after CD3 crosslinking. Overall, our work shows that the new LAT negative J.CaM1.7 cell subline could represent a new model to study the functions of the tyrosine kinase Lck and the LAT adaptor in TCR signaling, and their mutual interaction, which seems to constitute an essential early signaling event associated with the TCR/CD3 complex. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Early T Cell Signaling)
Show Figures

Figure 1

12 pages, 4386 KiB  
Article
A Multimodal Platform for Simultaneous T-cell Imaging, Defined Activation, and Mechanobiological Characterization
by Martin Fölser, Viktoria Motsch, René Platzer, Johannes B. Huppa and Gerhard J. Schütz
Cells 2021, 10(2), 235; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10020235 - 25 Jan 2021
Cited by 4 | Viewed by 3022
Abstract
T-cell antigen recognition is accompanied by extensive morphological rearrangements of the contact zone between the T-cell and the antigen-presenting cell (APC). This process involves binding of the T-cell receptor (TCR) complex to antigenic peptides presented via MHC on the APC surface, the interaction [...] Read more.
T-cell antigen recognition is accompanied by extensive morphological rearrangements of the contact zone between the T-cell and the antigen-presenting cell (APC). This process involves binding of the T-cell receptor (TCR) complex to antigenic peptides presented via MHC on the APC surface, the interaction of costimulatory and adhesion proteins, remodeling of the actin cytoskeleton, and the initiation of downstream signaling processes such as the release of intracellular calcium. However, multiparametric time-resolved analysis of these processes is hampered by the difficulty in recording the different readout modalities at high quality in parallel. In this study, we present a platform for simultaneous quantification of TCR distribution via total internal reflection fluorescence microscopy, of intracellular calcium levels, and of T-cell-exerted forces via atomic force microscopy (AFM). In our method, AFM cantilevers were used to bring single T-cells into contact with the activating surface. We designed the platform specifically to enable the study of T-cell triggering via functionalized fluid-supported lipid bilayers, which represent a widely accepted model system to stimulate T-cells in an antigen-specific manner. In this paper, we showcase the possibilities of this platform using primary transgenic T-cells triggered specifically via their cognate antigen presented by MHCII. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Early T Cell Signaling)
Show Figures

Figure 1

Review

Jump to: Research

13 pages, 946 KiB  
Review
Quantitative Methodologies to Dissect Immune Cell Mechanobiology
by Veronika Pfannenstill, Aurélien Barbotin, Huw Colin-York and Marco Fritzsche
Cells 2021, 10(4), 851; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10040851 - 9 Apr 2021
Cited by 2 | Viewed by 4242
Abstract
Mechanobiology seeks to understand how cells integrate their biomechanics into their function and behavior. Unravelling the mechanisms underlying these mechanobiological processes is particularly important for immune cells in the context of the dynamic and complex tissue microenvironment. However, it remains largely unknown how [...] Read more.
Mechanobiology seeks to understand how cells integrate their biomechanics into their function and behavior. Unravelling the mechanisms underlying these mechanobiological processes is particularly important for immune cells in the context of the dynamic and complex tissue microenvironment. However, it remains largely unknown how cellular mechanical force generation and mechanical properties are regulated and integrated by immune cells, primarily due to a profound lack of technologies with sufficient sensitivity to quantify immune cell mechanics. In this review, we discuss the biological significance of mechanics for immune cells across length and time scales, and highlight several experimental methodologies for quantifying the mechanics of immune cells. Finally, we discuss the importance of quantifying the appropriate mechanical readout to accelerate insights into the mechanobiology of the immune response. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Early T Cell Signaling)
Show Figures

Figure 1

Back to TopTop