1
|
Ghosh S, Wagenknecht-Wiesner A, Desai S, Vyphuis J, Ramos MS, Grazul JL, Baird BA. The Synergy between Topography and Lipid Domains in the Plasma Membrane of Mast Cells Controls the Localization of Signaling Proteins and Facilitates their Coordinated Activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624791. [PMID: 39605335 PMCID: PMC11601610 DOI: 10.1101/2024.11.22.624791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Similar to T cells and B cells, mast cell surfaces are dominated by microvilli, and like these other immune cells we showed with microvillar cartography (MC) that key signaling proteins for RBL mast cells localize to these topographical features. Although stabilization of ordered lipid nanodomains around antigen-crosslinked IgE-FcεRI is known to facilitate necessary coupling with Lyn tyrosine kinase to initiate transmembrane signaling in these mast cells, the relationship of ordered-lipid nanodomains to membrane topography had not been determined. With nanoscale resolution provided by MC, SEM and co-localization probability (CP) analysis, we found that FcεRI and Lyn kinase are positioned exclusively on the microvilli of resting mast cells in separate nano-assemblies, and upon antigen-activation they merge into overlapping populations together with the LAT scaffold protein, accompanied by elongation and merger of microvilli into ridge-like ruffles. With selective lipid probes, we further found that ordered-lipid nanodomains preferentially occupy microvillar membranes, contrasting with localization of disordered lipids to flatter regions. With this proximity of signaling proteins and ordered lipid nanodomains in microvilli, the mast cells are poised to respond sensitively and efficiently to antigen but only in the presence of this stimulus. Use of a short chain ceramide to disrupt ordered-lipid regions of the plasma membrane and evaluation with MC, CP, and flow cytometry provided strong evidence that the microvillar selective localization of signaling proteins and lipid environments is facilitated by the interplay between ordered-lipid nanodomains and actin attachment proteins, ERM (ezrin, radixin, moesin) and cofilin.
Collapse
Affiliation(s)
- Shirsendu Ghosh
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Department of Chemistry, Gandhi Institute of Technology and Management, Hyderabad Campus, Rudraram, Telangana 502329, India
| | | | - Shriya Desai
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| | - Jada Vyphuis
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| | | | - John L. Grazul
- Cornell Center for Materials Research, Cornell University, Ithaca, NY 14853
| | - Barbara A. Baird
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| |
Collapse
|
2
|
Neve-Oz Y, Sherman E, Raveh B. Bayesian metamodeling of early T-cell antigen receptor signaling accounts for its nanoscale activation patterns. Front Immunol 2024; 15:1412221. [PMID: 39524449 PMCID: PMC11543436 DOI: 10.3389/fimmu.2024.1412221] [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: 04/04/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
T cells respond swiftly, specifically, sensitively, and robustly to cognate antigens presented on the surface of antigen presenting cells. Existing microscopic models capture various aspects of early T-cell antigen receptor (TCR) signaling at the molecular level. However, none of these models account for the totality of the data, impeding our understanding of early T-cell activation. Here, we study early TCR signaling using Bayesian metamodeling, an approach for systematically integrating multiple partial models into a metamodel of a complex system. We inform the partial models using multiple published super-resolution microscopy datasets. Collectively, these datasets describe the spatiotemporal organization, activity, interactions, and dynamics of TCR, CD45 and Lck signaling molecules in the early-forming immune synapse, and the concurrent membrane alterations. The resulting metamodel accounts for a distinct nanoscale dynamic pattern that could not be accounted for by any of the partial models on their own: a ring of phosphorylated TCR molecules, enriched at the periphery of early T cell contacts and confined by a proximal ring of CD45 molecules. The metamodel suggests this pattern results from limited activity range for the Lck molecules, acting as signaling messengers between kinetically-segregated TCR and CD45 molecules. We assessed the potential effect of Lck activity range on TCR phosphorylation and robust T cell activation for various pMHC:TCR association strengths, in the specific setting of an initial contact. We also inspected the impact of localized Lck inhibition via Csk recruitment to pTCRs, and that of splicing isoforms of CD45 on kinetic segregation. Due to the inherent scalability and adaptability of integrating independent partial models via Bayesian metamodeling, this approach can elucidate additional aspects of cell signaling and decision making.
Collapse
Affiliation(s)
- Yair Neve-Oz
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| | - Barak Raveh
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
3
|
Li X, Chou T. Reliable ligand discrimination in stochastic multistep kinetic proofreading: First passage time vs. product counting strategies. PLoS Comput Biol 2024; 20:e1012183. [PMID: 38857304 PMCID: PMC11192422 DOI: 10.1371/journal.pcbi.1012183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/21/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
Cellular signaling, crucial for biological processes like immune response and homeostasis, relies on specificity and fidelity in signal transduction to accurately respond to stimuli amidst biological noise. Kinetic proofreading (KPR) is a key mechanism enhancing signaling specificity through time-delayed steps, although its effectiveness is debated due to intrinsic noise potentially reducing signal fidelity. In this study, we reformulate the theory of kinetic proofreading (KPR) by convolving multiple intermediate states into a single state and then define an overall "processing" time required to traverse these states. This simplification allows us to succinctly describe kinetic proofreading in terms of a single waiting time parameter, facilitating a more direct evaluation and comparison of KPR performance across different biological contexts such as DNA replication and T cell receptor (TCR) signaling. We find that loss of fidelity for longer proofreading steps relies on the specific strategy of information extraction and show that in the first-passage time (FPT) discrimination strategy, longer proofreading steps can exponentially improve the accuracy of KPR at the cost of speed. Thus, KPR can still be an effective discrimination mechanism in the high noise regime. However, in a product concentration-based discrimination strategy, longer proofreading steps do not necessarily lead to an increase in performance. However, by introducing activation thresholds on product concentrations, can we decompose the product-based strategy into a series of FPT-based strategies to better resolve the subtleties of KPR-mediated product discrimination. Our findings underscore the importance of understanding KPR in the context of how information is extracted and processed in the cell.
Collapse
Affiliation(s)
- Xiangting Li
- Department of Computational Medicine, University of California, Los Angeles, California, United States of America
| | - Tom Chou
- Department of Computational Medicine, University of California, Los Angeles, California, United States of America
- Department of Mathematics, University of California, Los Angeles, California, United States of America
| |
Collapse
|
4
|
Querol Cano L, Dunlock VME, Schwerdtfeger F, van Spriel AB. Membrane organization by tetraspanins and galectins shapes lymphocyte function. Nat Rev Immunol 2024; 24:193-212. [PMID: 37758850 DOI: 10.1038/s41577-023-00935-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 09/29/2023]
Abstract
Immune receptors are not randomly distributed at the plasma membrane of lymphocytes but are segregated into specialized domains that function as platforms to initiate signalling, as exemplified by the B cell or T cell receptor complex and the immunological synapse. 'Membrane-organizing proteins' and, in particular, tetraspanins and galectins, are crucial for controlling the spatiotemporal organization of immune receptors and other signalling proteins. Deficiencies in specific tetraspanins and galectins result in impaired immune synapse formation, lymphocyte proliferation, antibody production and migration, which can lead to impaired immunity, tumour development and autoimmunity. In contrast to conventional ligand-receptor interactions, membrane organizers interact in cis (on the same cell) and modulate receptor clustering, receptor dynamics and intracellular signalling. New findings have uncovered their complex and dynamic nature, revealing shared binding partners and collaborative activity in determining the composition of membrane domains. Therefore, immune receptors should not be envisaged as independent entities and instead should be studied in the context of their spatial organization in the lymphocyte membrane. We advocate for a novel approach to study lymphocyte function by globally analysing the role of membrane organizers in the assembly of different membrane complexes and discuss opportunities to develop therapeutic approaches that act via the modulation of membrane organization.
Collapse
Affiliation(s)
- Laia Querol Cano
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vera-Marie E Dunlock
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fabian Schwerdtfeger
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemiek B van Spriel
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
5
|
Acuto O. T-cell virtuosity in ''knowing thyself". Front Immunol 2024; 15:1343575. [PMID: 38415261 PMCID: PMC10896960 DOI: 10.3389/fimmu.2024.1343575] [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: 11/23/2023] [Accepted: 01/17/2024] [Indexed: 02/29/2024] Open
Abstract
Major Histocompatibility Complex (MHC) I and II and the αβ T-cell antigen receptor (TCRαβ) govern fundamental traits of adaptive immunity. They form a membrane-borne ligand-receptor system weighing host proteome integrity to detect contamination by nonself proteins. MHC-I and -II exhibit the "MHC-fold", which is able to bind a large assortment of short peptides as proxies for self and nonself proteins. The ensuing varying surfaces are mandatory ligands for Ig-like TCRαβ highly mutable binding sites. Conserved molecular signatures guide TCRαβ ligand binding sites to focus on the MHC-fold (MHC-restriction) while leaving many opportunities for its most hypervariable determinants to contact the peptide. This riveting molecular strategy affords many options for binding energy compatible with specific recognition and signalling aimed to eradicated microbial pathogens and cancer cells. While the molecular foundations of αβ T-cell adaptive immunity are largely understood, uncertainty persists on how peptide-MHC binding induces the TCRαβ signals that instruct cell-fate decisions. Solving this mystery is another milestone for understanding αβ T-cells' self/nonself discrimination. Recent developments revealing the innermost links between TCRαβ structural dynamics and signalling modality should help dissipate this long-sought-after enigma.
Collapse
Affiliation(s)
- Oreste Acuto
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Wither MJ, White WL, Pendyala S, Leanza PJ, Fowler DM, Kueh HY. Antigen perception in T cells by long-term Erk and NFAT signaling dynamics. Proc Natl Acad Sci U S A 2023; 120:e2308366120. [PMID: 38113261 PMCID: PMC10756264 DOI: 10.1073/pnas.2308366120] [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: 05/29/2023] [Accepted: 10/20/2023] [Indexed: 12/21/2023] Open
Abstract
Immune system threat detection hinges on T cells' ability to perceive varying peptide-major histocompatibility complex (pMHC) antigens. As the Erk and NFAT pathways link T cell receptor engagement to gene regulation, their signaling dynamics may convey information about pMHC inputs. To test this idea, we developed a dual reporter mouse strain and a quantitative imaging assay that, together, enable simultaneous monitoring of Erk and NFAT dynamics in live T cells over day-long timescales as they respond to varying pMHC inputs. Both pathways initially activate uniformly across various pMHC inputs but diverge only over longer (9+ h) timescales, enabling independent encoding of pMHC affinity and dose. These late signaling dynamics are decoded via multiple temporal and combinatorial mechanisms to generate pMHC-specific transcriptional responses. Our findings underscore the importance of long timescale signaling dynamics in antigen perception and establish a framework for understanding T cell responses under diverse contexts.
Collapse
Affiliation(s)
- Matthew J. Wither
- University of Washington, Department of Bioengineering, Seattle, WA98195
| | - William L. White
- University of Washington, Department of Bioengineering, Seattle, WA98195
| | - Sriram Pendyala
- University of Washington, Department of Genome Sciences, Seattle, WA98195
| | - Paul J. Leanza
- University of Washington, Department of Bioengineering, Seattle, WA98195
| | - Douglas M. Fowler
- University of Washington, Department of Genome Sciences, Seattle, WA98195
| | - Hao Yuan Kueh
- University of Washington, Department of Bioengineering, Seattle, WA98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA98109
| |
Collapse
|
7
|
Dustin ML. Recent advances in understanding TCR signaling: a synaptic perspective. Fac Rev 2023; 12:25. [PMID: 37900153 PMCID: PMC10608137 DOI: 10.12703/r/12-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023] Open
Abstract
The T cell receptor is a multi-subunit complex that carries out a range of recognition tasks for multiple lymphocyte types and translates recognition into signals that regulate survival, growth, differentiation, and effector functions for innate and adaptive host defense. Recent advances include the cryo-electron microscopy-based structure of the extracellular and transmembrane components of the complex, new information about coupling to intracellular partners, lateral associations in the membrane that all add to our picture of the T cell signaling machinery, and how signal termination relates to effector function. This review endeavors to integrate structural and biochemical information through the lens of the immunological synapse- the critical interface with the antigen-presenting cell.
Collapse
Affiliation(s)
- Michael L Dustin
- Kennedy Institute of Rheumatology, The University of Oxford, Oxford, UK
| |
Collapse
|
8
|
Abstract
T cell activation is initiated by the recognition of specific antigenic peptides and subsequently accomplished by complex signaling cascades. These aspects have been extensively studied for decades as pivotal factors in the establishment of adaptive immunity. However, how receptors or signaling molecules are organized in the resting state prior to encountering antigens has received less attention. Recent advancements in super-resolution microscopy techniques have revealed topographically controlled pre-formed organization of key molecules involved in antigen recognition and signal transduction on microvillar projections of T cells before activation and substantial effort has been dedicated to characterizing the topological structure of resting T cells over the past decade. This review will summarize our current understanding of how key surface receptors are pre-organized on the T-cell plasma membrane and discuss the potential role of these receptors, which are preassembled prior to ligand binding in the early activation events of T cells.
Collapse
Affiliation(s)
- Yunmin Jung
- Department of Nano-Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science, Seoul, Republic of Korea
| |
Collapse
|
9
|
Morgan J, Lindsay AE. Modulation of antigen discrimination by duration of immune contacts in a kinetic proofreading model of T cell activation with extreme statistics. PLoS Comput Biol 2023; 19:e1011216. [PMID: 37647345 PMCID: PMC10497171 DOI: 10.1371/journal.pcbi.1011216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/12/2023] [Accepted: 08/05/2023] [Indexed: 09/01/2023] Open
Abstract
T cells form transient cell-to-cell contacts with antigen presenting cells (APCs) to facilitate surface interrogation by membrane bound T cell receptors (TCRs). Upon recognition of molecular signatures (antigen) of pathogen, T cells may initiate an adaptive immune response. The duration of the T cell/APC contact is observed to vary widely, yet it is unclear what constructive role, if any, such variations might play in immune signaling. Modeling efforts describing antigen discrimination often focus on steady-state approximations and do not account for the transient nature of cellular contacts. Within the framework of a kinetic proofreading (KP) mechanism, we develop a stochastic First Receptor Activation Model (FRAM) describing the likelihood that a productive immune signal is produced before the expiry of the contact. Through the use of extreme statistics, we characterize the probability that the first TCR triggering is induced by a rare agonist antigen and not by that of an abundant self-antigen. We show that defining positive immune outcomes as resilience to extreme statistics and sensitivity to rare events mitigates classic tradeoffs associated with KP. By choosing a sufficient number of KP steps, our model is able to yield single agonist sensitivity whilst remaining non-reactive to large populations of self antigen, even when self and agonist antigen are similar in dissociation rate to the TCR but differ largely in expression. Additionally, our model achieves high levels of accuracy even when agonist positive APCs encounters are rare. Finally, we discuss potential biological costs associated with high classification accuracy, particularly in challenging T cell environments.
Collapse
Affiliation(s)
- Jonathan Morgan
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, South Bend, Indiana, United States of America
- Biophysics Graduate Program, University of Notre Dame, South Bend, Indiana, United States of America
| | - Alan E. Lindsay
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, South Bend, Indiana, United States of America
| |
Collapse
|
10
|
Wither MJ, White WL, Pendyala S, Leanza PJ, Fowler D, Kueh HY. Antigen perception in T cells by long-term Erk and NFAT signaling dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543260. [PMID: 37333368 PMCID: PMC10274683 DOI: 10.1101/2023.06.01.543260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Immune system threat detection hinges on T cells' ability to perceive varying peptide major-histocompatibility complex (pMHC) antigens. As the Erk and NFAT pathways link T cell receptor engagement to gene regulation, their signaling dynamics may convey information about pMHC inputs. To test this idea, we developed a dual reporter mouse strain and a quantitative imaging assay that, together, enable simultaneous monitoring of Erk and NFAT dynamics in live T cells over day-long timescales as they respond to varying pMHC inputs. Both pathways initially activate uniformly across various pMHC inputs, but diverge only over longer (9+ hrs) timescales, enabling independent encoding of pMHC affinity and dose. These late signaling dynamics are decoded via multiple temporal and combinatorial mechanisms to generate pMHC-specific transcriptional responses. Our findings underscore the importance of long timescale signaling dynamics in antigen perception, and establish a framework for understanding T cell responses under diverse contexts. SIGNIFICANCE STATEMENT To counter diverse pathogens, T cells mount distinct responses to varying peptide-major histocompatibility complex ligands (pMHCs). They perceive the affinity of pMHCs for the T cell receptor (TCR), which reflects its foreignness, as well as pMHC abundance. By tracking signaling responses in single living cells to different pMHCs, we find that T cells can independently perceive pMHC affinity vs dose, and encode this information through the dynamics of Erk and NFAT signaling pathways downstream of the TCR. These dynamics are jointly decoded by gene regulatory mechanisms to produce pMHC-specific activation responses. Our work reveals how T cells can elicit tailored functional responses to diverse threats and how dysregulation of these responses may lead to immune pathologies.
Collapse
|
11
|
Hyun J, Kim SJ, Cho SD, Kim HW. Mechano-modulation of T cells for cancer immunotherapy. Biomaterials 2023; 297:122101. [PMID: 37023528 DOI: 10.1016/j.biomaterials.2023.122101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/12/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Immunotherapy, despite its promise for future anti-cancer approach, faces significant challenges, such as off-tumor side effects, innate or acquired resistance, and limited infiltration of immune cells into stiffened extracellular matrix (ECM). Recent studies have highlighted the importance of mechano-modulation/-activation of immune cells (mainly T cells) for effective caner immunotherapy. Immune cells are highly sensitive to the applied physical forces and matrix mechanics, and reciprocally shape the tumor microenvironment. Engineering T cells with tuned properties of materials (e.g., chemistry, topography, and stiffness) can improve their expansion and activation ex vivo, and their ability to mechano-sensing the tumor specific ECM in vivo where they perform cytotoxic effects. T cells can also be exploited to secrete enzymes that soften ECM, thus increasing tumor infiltration and cellular therapies. Furthermore, T cells, such as chimeric antigen receptor (CAR)-T cells, genomic engineered to be spatiotemporally controllable by physical stimuli (e.g., ultrasound, heat, or light), can mitigate adverse off-tumor effects. In this review, we communicate these recent cutting-edge endeavors devoted to mechano-modulating/-activating T cells for effective cancer immunotherapy, and discuss future prospects and challenges in this field.
Collapse
|
12
|
Jenkins E, Körbel M, O'Brien-Ball C, McColl J, Chen KY, Kotowski M, Humphrey J, Lippert AH, Brouwer H, Santos AM, Lee SF, Davis SJ, Klenerman D. Antigen discrimination by T cells relies on size-constrained microvillar contact. Nat Commun 2023; 14:1611. [PMID: 36959206 PMCID: PMC10036606 DOI: 10.1038/s41467-023-36855-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 02/21/2023] [Indexed: 03/25/2023] Open
Abstract
T cells use finger-like protrusions called 'microvilli' to interrogate their targets, but why they do so is unknown. To form contacts, T cells must overcome the highly charged, barrier-like layer of large molecules forming a target cell's glycocalyx. Here, T cells are observed to use microvilli to breach a model glycocalyx barrier, forming numerous small (<0.5 μm diameter) contacts each of which is stabilized by the small adhesive protein CD2 expressed by the T cell, and excludes large proteins including CD45, allowing sensitive, antigen dependent TCR signaling. In the absence of the glycocalyx or when microvillar contact-size is increased by enhancing CD2 expression, strong signaling occurs that is no longer antigen dependent. Our observations suggest that, modulated by the opposing effects of the target cell glycocalyx and small adhesive proteins, the use of microvilli equips T cells with the ability to effect discriminatory receptor signaling.
Collapse
Affiliation(s)
- Edward Jenkins
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Markus Körbel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Caitlin O'Brien-Ball
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - James McColl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Kevin Y Chen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Mateusz Kotowski
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Jane Humphrey
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Anna H Lippert
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Heather Brouwer
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Steven F Lee
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
13
|
Saltukoglu D, Özdemir B, Holtmannspötter M, Reski R, Piehler J, Kurre R, Reth M. Plasma membrane topography governs the 3D dynamic localization of IgM B cell antigen receptor clusters. EMBO J 2023; 42:e112030. [PMID: 36594262 PMCID: PMC9929642 DOI: 10.15252/embj.2022112030] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 01/04/2023] Open
Abstract
B lymphocytes recognize bacterial or viral antigens via different classes of the B cell antigen receptor (BCR). Protrusive structures termed microvilli cover lymphocyte surfaces, and are thought to perform sensory functions in screening antigen-bearing surfaces. Here, we have used lattice light-sheet microscopy in combination with tailored custom-built 4D image analysis to study the cell-surface topography of B cells of the Ramos Burkitt's Lymphoma line and the spatiotemporal organization of the IgM-BCR. Ramos B-cell surfaces were found to form dynamic networks of elevated ridges bridging individual microvilli. A fraction of membrane-localized IgM-BCR was found in clusters, which were mainly associated with the ridges and the microvilli. The dynamic ridge-network organization and the IgM-BCR cluster mobility were linked, and both were controlled by Arp2/3 complex activity. Our results suggest that dynamic topographical features of the cell surface govern the localization and transport of IgM-BCR clusters to facilitate antigen screening by B cells.
Collapse
Affiliation(s)
- Deniz Saltukoglu
- Department of Molecular Immunology, Biology III, Faculty of BiologyUniversity of FreiburgFreiburgGermany
- Signaling Research Centers CIBSS and BIOSSUniversity of FreiburgFreiburgGermany
| | - Bugra Özdemir
- Signaling Research Centers CIBSS and BIOSSUniversity of FreiburgFreiburgGermany
- Plant Biotechnology, Faculty of BiologyUniversity of FreiburgFreiburgGermany
- Present address:
Euro‐BioImaging, European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Michael Holtmannspötter
- Department of Biology/Chemistry and Center for Cellular NanoanalyticsOsnabrück UniversityOsnabrückGermany
| | - Ralf Reski
- Signaling Research Centers CIBSS and BIOSSUniversity of FreiburgFreiburgGermany
- Plant Biotechnology, Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Jacob Piehler
- Department of Biology/Chemistry and Center for Cellular NanoanalyticsOsnabrück UniversityOsnabrückGermany
| | - Rainer Kurre
- Department of Biology/Chemistry and Center for Cellular NanoanalyticsOsnabrück UniversityOsnabrückGermany
| | - Michael Reth
- Department of Molecular Immunology, Biology III, Faculty of BiologyUniversity of FreiburgFreiburgGermany
- Signaling Research Centers CIBSS and BIOSSUniversity of FreiburgFreiburgGermany
| |
Collapse
|
14
|
Morgan J, Pettmann J, Dushek O, Lindsay AE. T cell microvilli simulations show operation near packing limit and impact on antigen recognition. Biophys J 2022; 121:4128-4136. [PMID: 36181267 PMCID: PMC9675027 DOI: 10.1016/j.bpj.2022.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 07/11/2022] [Accepted: 09/26/2022] [Indexed: 11/02/2022] Open
Abstract
T cells are immune cells that continuously scan for foreign-derived antigens on the surfaces of nearly all cells, termed antigen-presenting cells (APCs). They do this by dynamically extending numerous protrusions called microvilli (MVs) that contain T cell receptors toward the APC surface in order to scan for antigens. The number, size, and dynamics of these MVs, and the complex multiscale topography that results, play a yet unknown role in antigen recognition. We develop an anatomically informed model that confines antigen recognition to small areas representing MVs that can dynamically form and dissolve and use the model to study how MV dynamics impact antigen sensitivity and discrimination. We find that MV surveillance reduces antigen sensitivity compared with a completely flat interface, unless MV are stabilized in an antigen-dependent manner, and observe that MVs have only a modest impact on antigen discrimination. The model highlights that MV contacts optimize the competing demands of fast scanning speeds of the APC surface with antigen sensitivity. Our model predicts an interface packing fraction that corresponds closely to those observed experimentally, indicating that T cells operate their MVs near the limits imposed by anatomical and geometric constraints. Finally, we find that observed MV contact lifetimes can be largely influenced by conditions in the T cell/APC interface, with these lifetimes often being longer than the simulation or experimental observation period. This work highlights the role of MVs in antigen recognition.
Collapse
Affiliation(s)
- Jonathan Morgan
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, Indiana; Biophysics Graduate Program, University of Notre Dame, Notre Dame, Indiana
| | - Johannes Pettmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Alan E Lindsay
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, Indiana.
| |
Collapse
|
15
|
Sanders EW, Carr AR, Bruggeman E, Körbel M, Benaissa SI, Donat RF, Santos AM, McColl J, O'Holleran K, Klenerman D, Davis SJ, Lee SF, Ponjavic A. resPAINT: Accelerating Volumetric Super-Resolution Localisation Microscopy by Active Control of Probe Emission. Angew Chem Int Ed Engl 2022; 61:e202206919. [PMID: 35876263 DOI: 10.1002/anie.202206919] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Indexed: 01/07/2023]
Abstract
Points for accumulation in nanoscale topography (PAINT) allows practically unlimited measurements in localisation microscopy but is limited by background fluorescence at high probe concentrations, especially in volumetric imaging. We present reservoir-PAINT (resPAINT), which combines PAINT and active control of probe photophysics. In resPAINT, an activatable probe "reservoir" accumulates on target, enabling a 50-fold increase in localisation rate versus conventional PAINT, without compromising contrast. By combining resPAINT with large depth-of-field microscopy, we demonstrate super-resolution imaging of entire cell surfaces. We generalise the approach by implementing various switching strategies and 3D imaging techniques. Finally, we use resPAINT with a Fab to image membrane proteins, extending the operating regime of PAINT to include a wider range of biological interactions.
Collapse
Affiliation(s)
- Edward W Sanders
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Alexander R Carr
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Ezra Bruggeman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Markus Körbel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Sarah I Benaissa
- Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, CB2 3DY, UK
| | - Robert F Donat
- Radcliffe Department of Medicine and United Kingdom Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana M Santos
- Radcliffe Department of Medicine and United Kingdom Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - James McColl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Kevin O'Holleran
- Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, CB2 3DY, UK
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Simon J Davis
- Radcliffe Department of Medicine and United Kingdom Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Steven F Lee
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Aleks Ponjavic
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- School of Physics and Astronomy, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
- School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| |
Collapse
|
16
|
Sanders EW, Carr AR, Bruggeman E, Körbel M, Benaissa SI, Donat RF, Santos AM, McColl J, O'Holleran K, Klenerman D, Davis SJ, Lee SF, Ponjavic A. resPAINT: Accelerating Volumetric Super-Resolution Localisation Microscopy by Active Control of Probe Emission. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202206919. [PMID: 38505515 PMCID: PMC10946633 DOI: 10.1002/ange.202206919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Indexed: 03/21/2024]
Abstract
Points for accumulation in nanoscale topography (PAINT) allows practically unlimited measurements in localisation microscopy but is limited by background fluorescence at high probe concentrations, especially in volumetric imaging. We present reservoir-PAINT (resPAINT), which combines PAINT and active control of probe photophysics. In resPAINT, an activatable probe "reservoir" accumulates on target, enabling a 50-fold increase in localisation rate versus conventional PAINT, without compromising contrast. By combining resPAINT with large depth-of-field microscopy, we demonstrate super-resolution imaging of entire cell surfaces. We generalise the approach by implementing various switching strategies and 3D imaging techniques. Finally, we use resPAINT with a Fab to image membrane proteins, extending the operating regime of PAINT to include a wider range of biological interactions.
Collapse
Affiliation(s)
- Edward W. Sanders
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
| | - Alexander R. Carr
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
| | - Ezra Bruggeman
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
| | - Markus Körbel
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
| | - Sarah I. Benaissa
- Cambridge Advanced Imaging CentreUniversity of CambridgeCambridgeCB2 3DYUK
| | - Robert F. Donat
- Radcliffe Department of Medicine and United Kingdom Medical Research Council Human Immunology UnitJohn Radcliffe HospitalUniversity of OxfordOxfordOX3 9DSUK
| | - Ana M. Santos
- Radcliffe Department of Medicine and United Kingdom Medical Research Council Human Immunology UnitJohn Radcliffe HospitalUniversity of OxfordOxfordOX3 9DSUK
| | - James McColl
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
| | - Kevin O'Holleran
- Cambridge Advanced Imaging CentreUniversity of CambridgeCambridgeCB2 3DYUK
| | - David Klenerman
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
| | - Simon J. Davis
- Radcliffe Department of Medicine and United Kingdom Medical Research Council Human Immunology UnitJohn Radcliffe HospitalUniversity of OxfordOxfordOX3 9DSUK
| | - Steven F. Lee
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
| | - Aleks Ponjavic
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
- School of Physics and AstronomyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of Food Science and NutritionUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| |
Collapse
|
17
|
Göhring J, Schrangl L, Schütz GJ, Huppa JB. Mechanosurveillance: Tiptoeing T Cells. Front Immunol 2022; 13:886328. [PMID: 35693808 PMCID: PMC9178122 DOI: 10.3389/fimmu.2022.886328] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/19/2022] [Indexed: 11/28/2022] Open
Abstract
Efficient scanning of tissue that T cells encounter during their migratory life is pivotal to protective adaptive immunity. In fact, T cells can detect even a single antigenic peptide/MHC complex (pMHC) among thousands of structurally similar yet non-stimulatory endogenous pMHCs on the surface of antigen-presenting cells (APCs) or target cells. Of note, the glycocalyx of target cells, being composed of proteoglycans and bulky proteins, is bound to affect and even modulate antigen recognition by posing as a physical barrier. T cell-resident microvilli are actin-rich membrane protrusions that puncture through such barriers and thereby actively place the considerably smaller T-cell antigen receptors (TCRs) in close enough proximity to APC-presented pMHCs so that productive interactions may occur efficiently yet under force. We here review our current understanding of how the plasticity of T-cell microvilli and physicochemical properties of the glycocalyx may affect early events in T-cell activation. We assess insights gained from studies on T-cell plasma membrane ultrastructure and provide an update on current efforts to integrate biophysical aspects such as the amplitude and directionality of TCR-imposed mechanical forces and the distribution and lateral mobility of plasma membrane-resident signaling molecules into a more comprehensive view on sensitized T-cell antigen recognition.
Collapse
Affiliation(s)
- Janett Göhring
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Institute of Applied Physics, TU Wien, Vienna, Austria
- *Correspondence: Janett Göhring,
| | | | | | - Johannes B. Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Brameshuber M, Klotzsch E, Ponjavic A, Sezgin E. Understanding immune signaling using advanced imaging techniques. Biochem Soc Trans 2022; 50:853-866. [PMID: 35343569 PMCID: PMC9162467 DOI: 10.1042/bst20210479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022]
Abstract
Advanced imaging is key for visualizing the spatiotemporal regulation of immune signaling which is a complex process involving multiple players tightly regulated in space and time. Imaging techniques vary in their spatial resolution, spanning from nanometers to micrometers, and in their temporal resolution, ranging from microseconds to hours. In this review, we summarize state-of-the-art imaging methodologies and provide recent examples on how they helped to unravel the mysteries of immune signaling. Finally, we discuss the limitations of current technologies and share our insights on how to overcome these limitations to visualize immune signaling with unprecedented fidelity.
Collapse
Affiliation(s)
- Mario Brameshuber
- Institute of Applied Physics – Biophysics, TU Wien, 1040 Vienna, Austria
| | - Enrico Klotzsch
- Humboldt-Universität zu Berlin, Institut für Biophysik, Experimentelle Biophysik Mechanobiologie, Sitz Invalidenstrasse 42, 10115 Berlin, Germany
| | - Aleks Ponjavic
- School of Physics and Astronomy, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
- School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| |
Collapse
|
19
|
Approach to map nanotopography of cell surface receptors. Commun Biol 2022; 5:218. [PMID: 35264712 PMCID: PMC8907216 DOI: 10.1038/s42003-022-03152-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/09/2022] [Indexed: 12/18/2022] Open
Abstract
Cells communicate with their environment via surface receptors, but nanoscopic receptor organization with respect to complex cell surface morphology remains unclear. This is mainly due to a lack of accessible, robust and high-resolution methods. Here, we present an approach for mapping the topography of receptors at the cell surface with nanometer precision. The method involves coating glass coverslips with glycine, which preserves the fine membrane morphology while allowing immobilized cells to be positioned close to the optical surface. We developed an advanced and simplified algorithm for the analysis of single-molecule localization data acquired in a biplane detection scheme. These advancements enable direct and quantitative mapping of protein distribution on ruffled plasma membranes with near isotropic 3D nanometer resolution. As demonstrated successfully for CD4 and CD45 receptors, the described workflow is a straightforward quantitative technique to study molecules and their interactions at the complex surface nanomorphology of differentiated metazoan cells. A super-resolution localisation-based method is shown to map receptor topography in immune cells, which allows quantitative study of molecules and their interactions at the complex surface nanomorphology of cells.
Collapse
|
20
|
Goyette J, Depoil D, Yang Z, Isaacson SA, Allard J, van der Merwe PA, Gaus K, Dustin ML, Dushek O. Dephosphorylation accelerates the dissociation of ZAP70 from the T cell receptor. Proc Natl Acad Sci U S A 2022; 119:e2116815119. [PMID: 35197288 PMCID: PMC8892339 DOI: 10.1073/pnas.2116815119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/11/2021] [Indexed: 11/20/2022] Open
Abstract
Protein-protein binding domains are critical in signaling networks. Src homology 2 (SH2) domains are binding domains that interact with sequences containing phosphorylated tyrosines. A subset of SH2 domain-containing proteins has tandem domains, which are thought to enhance binding affinity and specificity. However, a trade-off exists between long-lived binding and the ability to rapidly reverse signaling, which is a critical requirement of noise-filtering mechanisms such as kinetic proofreading. Here, we use modeling to show that the unbinding rate of tandem, but not single, SH2 domains can be accelerated by phosphatases. Using surface plasmon resonance, we show that the phosphatase CD45 can accelerate the unbinding rate of zeta chain-associated protein kinase 70 (ZAP70), a tandem SH2 domain-containing kinase, from biphosphorylated peptides from the T cell receptor (TCR). An important functional prediction of accelerated unbinding is that the intracellular ZAP70-TCR half-life in T cells will not be fixed but rather, dependent on the extracellular TCR-antigen half-life, and we show that this is the case in both cell lines and primary T cells. The work highlights that tandem SH2 domains can break the trade-off between signal fidelity (requiring long half-life) and signal reversibility (requiring short half-life), which is a key requirement for T cell antigen discrimination mediated by kinetic proofreading.
Collapse
Affiliation(s)
- Jesse Goyette
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney 2052, NSW, Australia;
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney 2052, NSW, Australia
| | - David Depoil
- The Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY Oxford, United Kingdom
| | - Zhengmin Yang
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney 2052, NSW, Australia
| | - Samuel A Isaacson
- Department of Mathematics and Statistics, Boston University, Boston, MA 02215
| | - Jun Allard
- Center for Complex Biological Systems, University of California, Irvine, CA 92697
| | - P Anton van der Merwe
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE Oxford, United Kingdom
| | - Katharina Gaus
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney 2052, NSW, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney 2052, NSW, Australia
| | - Michael L Dustin
- The Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY Oxford, United Kingdom;
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE Oxford, United Kingdom
| |
Collapse
|
21
|
Simulation of receptor triggering by kinetic segregation shows role of oligomers and close-contacts. Biophys J 2022; 121:1660-1674. [PMID: 35367423 PMCID: PMC9117938 DOI: 10.1016/j.bpj.2022.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 03/28/2022] [Indexed: 11/23/2022] Open
Abstract
The activation of T cells, key players of the immune system, involves local evacuation of phosphatase CD45 from a region of the T cell's surface, segregating it from the T cell receptor. What drives this evacuation? In the presence of antigen, what ensures evacuation happens in the subsecond timescales necessary to initiate signaling? In the absence of antigen, what mechanisms ensure that evacuation does not happen spontaneously, which could cause signaling errors? Phenomena known to influence spatial organization of CD45 or similar surface molecules include diffusive motion in the lipid bilayer, oligomerization reactions, and mechanical compression against a nearby surface, such as that of the cell presenting the antigen. Computer simulations can investigate hypothesized spatiotemporal mechanisms of T cell signaling. The challenge to computational studies of evacuation is that the base process, spontaneous evacuation by simple diffusion, is in the extreme rare event limit, meaning direct stochastic simulation is unfeasible. Here, we combine particle-based spatial stochastic simulation with the weighted ensemble method for rare events to compute the mean first passage time for cell surface availability by surface reorganization of CD45. We confirm mathematical estimates that, at physiological concentrations, spontaneous evacuation is extremely rare, roughly 300 years. We find that dimerization decreases the time required for evacuation. A weak bimolecular interaction (dissociation constant estimate 460 μM) is sufficient for an order of magnitude reduction of spontaneous evacuation times, and oligomerization to hexamers reduces times to below 1 s. This introduces a mechanism whereby externally induced CD45 oligomerization could significantly modify T cell function. For large regions of close contact, such as those induced by large microvilli, molecular size and compressibility imply a nonzero reentry probability of 60%, decreasing evacuation times. Simulations show that these reduced evacuation times are still unrealistically long (even with a fourfold variation centered around previous estimates of parameters), suggesting that a yet-to-be-described mechanism, besides compressional exclusion at a close contact, drives evacuation.
Collapse
|
22
|
Velas L, Brameshuber M, Huppa JB, Kurz E, Dustin ML, Zelger P, Jesacher A, Schütz GJ. Three-Dimensional Single Molecule Localization Microscopy Reveals the Topography of the Immunological Synapse at Isotropic Precision below 15 nm. NANO LETTERS 2021; 21:9247-9255. [PMID: 34709845 PMCID: PMC8587899 DOI: 10.1021/acs.nanolett.1c03160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
T-cells engage with antigen-presenting cells in search for antigenic peptides and form transient interfaces termed immunological synapses. Synapse topography affects receptor binding rates and the mutual segregation of proteins due to size exclusion effects. It is hence important to determine the 3D topography of the immunological synapse at high precision. Current methods provide only rather coarse images of the protein distribution within the synapse. Here, we applied supercritical angle fluorescence microscopy combined with defocused imaging, which allows three-dimensional single molecule localization microscopy (3D-SMLM) at an isotropic localization precision below 15 nm. Experiments were performed on hybrid synapses between primary T-cells and functionalized glass-supported lipid bilayers. We used 3D-SMLM to quantify the cleft size within the synapse by mapping the position of the T-cell receptor (TCR) with respect to the supported lipid bilayer, yielding average distances of 18 nm up to 31 nm for activating and nonactivating bilayers, respectively.
Collapse
Affiliation(s)
- Lukas Velas
- Institute
of Applied Physics, TU Wien, 1040 Vienna, Austria
| | | | - Johannes B. Huppa
- Institute
for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology
and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Elke Kurz
- Kennedy
Institute of Rheumatology, University of
Oxford, OX3 7FY Oxford, United Kingdom
| | - Michael L. Dustin
- Kennedy
Institute of Rheumatology, University of
Oxford, OX3 7FY Oxford, United Kingdom
| | - Philipp Zelger
- Division
for Biomedical Physics, Medical University
of Innsbruck, 6020 Innsbruck, Austria
| | - Alexander Jesacher
- Division
for Biomedical Physics, Medical University
of Innsbruck, 6020 Innsbruck, Austria
| | | |
Collapse
|
23
|
Trapping or slowing the diffusion of T cell receptors at close contacts initiates T cell signaling. Proc Natl Acad Sci U S A 2021; 118:2024250118. [PMID: 34526387 PMCID: PMC8488633 DOI: 10.1073/pnas.2024250118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 12/27/2022] Open
Abstract
T cell activation is initiated by T cell receptor (TCR) phosphorylation. This requires the local depletion of large receptor-type phosphatases from "close contacts" formed when T cells interact with surfaces presenting agonistic TCR ligands, but exactly how the ligands potentiate signaling is unclear. It has been proposed that TCR ligands could enhance receptor phosphorylation and signaling just by holding TCRs in phosphatase-depleted close contacts, but this has not been directly tested. We devised simple methods to move the TCR in and out of close contacts formed by T cells interacting with supported lipid bilayers (SLBs) and to slow the receptor's diffusion in the contacts, using a series of anti-CD3ε Fab- and ligand-based adducts of the receptor. TCRs engaging a Fab extended with the large extracellular region of CD45 were excluded from contacts and produced no signaling. Conversely, allowing the extended Fab to become tethered to the SLB trapped the TCR in the close contacts, leading to very strong signaling. Importantly, attaching untethered anti-CD3ε Fab or peptide/MHC ligands, each of which were largely inactive in solution but both of which reduced TCR diffusion in close contacts approximately fivefold, also initiated signaling during cell/SLB contact. Our findings indicate that holding TCRs in close contacts or simply slowing their diffusion in phosphatase-depleted regions of the cell surface suffices to initiate signaling, effects we could reproduce in single-particle stochastic simulations. Our study shows that the TCR is preconfigured for signaling in a way that allows it to be triggered by ligands acting simply as receptor "traps."
Collapse
|
24
|
Wilhelm KB, Morita S, McAffee DB, Kim S, O'Dair MK, Groves JT. Height, but not binding epitope, affects the potency of synthetic TCR agonists. Biophys J 2021; 120:3869-3880. [PMID: 34453921 PMCID: PMC8511163 DOI: 10.1016/j.bpj.2021.08.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/27/2022] Open
Abstract
Under physiological conditions, peptide-major histocompatibility complex (pMHC) molecules can trigger T cell receptors (TCRs) as monovalent ligands that are sparsely distributed on the plasma membrane of an antigen-presenting cell. TCRs can also be triggered by artificial clustering, such as with pMHC tetramers or antibodies; however, these strategies circumvent many of the natural ligand discrimination mechanisms of the T cell and can elicit nonphysiological signaling activity. We have recently introduced a synthetic TCR agonist composed of an anti-TCRβ Fab′ antibody fragment covalently bound to a DNA oligonucleotide, which serves as a membrane anchor. This Fab′-DNA ligand efficiently triggers TCR as a monomer when membrane associated and exhibits a potency and activation profile resembling agonist pMHC. In this report, we explore the geometric requirements for efficient TCR triggering and cellular activation by Fab′-DNA ligands. We find that T cells are insensitive to the ligand binding epitope on the TCR complex but that length of the DNA tether is important. Increasing, the intermembrane distance spanned by Fab′-DNA:TCR complexes decreases TCR triggering efficiency and T cell activation potency, consistent with the kinetic-segregation model of TCR triggering. These results establish design parameters for constructing synthetic TCR agonists that are able to activate polyclonal T cell populations, such as T cells from a human patient, in a similar manner as the native pMHC ligand.
Collapse
Affiliation(s)
- Kiera B Wilhelm
- Department of Chemistry, University of California, Berkeley, California
| | - Shumpei Morita
- Department of Chemistry, University of California, Berkeley, California
| | - Darren B McAffee
- Department of Chemistry, University of California, Berkeley, California
| | - Sungi Kim
- Department of Chemistry, University of California, Berkeley, California
| | - Mark K O'Dair
- Department of Chemistry, University of California, Berkeley, California
| | - Jay T Groves
- Department of Chemistry, University of California, Berkeley, California.
| |
Collapse
|
25
|
Urbančič I, Schiffelers L, Jenkins E, Gong W, Santos AM, Schneider F, O'Brien-Ball C, Vuong MT, Ashman N, Sezgin E, Eggeling C. Aggregation and mobility of membrane proteins interplay with local lipid order in the plasma membrane of T cells. FEBS Lett 2021; 595:2127-2146. [PMID: 34160065 DOI: 10.1002/1873-3468.14153] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/07/2021] [Accepted: 06/18/2021] [Indexed: 01/17/2023]
Abstract
To disentangle the elusive lipid-protein interactions in T-cell activation, we investigate how externally imposed variations in mobility of key membrane proteins (T-cell receptor [TCR], kinase Lck, and phosphatase CD45) affect the local lipid order and protein colocalisation. Using spectral imaging with polarity-sensitive membrane probes in model membranes and live Jurkat T cells, we find that partial immobilisation of proteins (including TCR) by aggregation or ligand binding changes their preference towards a more ordered lipid environment, which can recruit Lck. Our data suggest that the cellular membrane is poised to modulate the frequency of protein encounters upon alterations of their mobility, for example in ligand binding, which offers new mechanistic insight into the involvement of lipid-mediated interactions in membrane-hosted signalling events.
Collapse
Affiliation(s)
- Iztok Urbančič
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
- Jožef Stefan Institute, Ljubljana, Slovenia
| | - Lisa Schiffelers
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Edward Jenkins
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Weijian Gong
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Ana Mafalda Santos
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Falk Schneider
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | | | - Mai Tuyet Vuong
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Nicole Ashman
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Erdinc Sezgin
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
- Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Christian Eggeling
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
- Institute of Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Germany
- Leibniz Institute of Photonic Technology e.V., Jena, Germany
| |
Collapse
|
26
|
Jung Y, Wen L, Altman A, Ley K. CD45 pre-exclusion from the tips of T cell microvilli prior to antigen recognition. Nat Commun 2021; 12:3872. [PMID: 34162836 PMCID: PMC8222282 DOI: 10.1038/s41467-021-23792-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
The tyrosine phosphatase CD45 is a major gatekeeper for restraining T cell activation. Its exclusion from the immunological synapse (IS) is crucial for T cell receptor (TCR) signal transduction. Here, we use expansion super-resolution microscopy to reveal that CD45 is mostly pre-excluded from the tips of microvilli (MV) on primary T cells prior to antigen encounter. This pre-exclusion is diminished by depleting cholesterol or by engineering the transmembrane domain of CD45 to increase its membrane integration length, but is independent of the CD45 extracellular domain. We further show that brief MV-mediated contacts can induce Ca2+ influx in mouse antigen-specific T cells engaged by antigen-pulsed antigen presenting cells (APC). We propose that the scarcity of CD45 phosphatase activity at the tips of MV enables or facilitates TCR triggering from brief T cell-APC contacts before formation of a stable IS, and that these MV-mediated contacts represent the earliest step in the initiation of a T cell adaptive immune response.
Collapse
Affiliation(s)
- Yunmin Jung
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | - Lai Wen
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Amnon Altman
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Klaus Ley
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
27
|
Souissi M, Pernier J, Rossier O, Giannone G, Le Clainche C, Helfer E, Sengupta K. Integrin-Functionalised Giant Unilamellar Vesicles via Gel-Assisted Formation: Good Practices and Pitfalls. Int J Mol Sci 2021; 22:6335. [PMID: 34199292 PMCID: PMC8231826 DOI: 10.3390/ijms22126335] [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: 04/28/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 01/16/2023] Open
Abstract
Giant unilamellar vesicles (GUV) are powerful tools to explore physics and biochemistry of the cell membrane in controlled conditions. For example, GUVs were extensively used to probe cell adhesion, but often using non-physiological linkers, due to the difficulty of incorporating transmembrane adhesion proteins into model membranes. Here we describe a new protocol for making GUVs incorporating the transmembrane protein integrin using gel-assisted swelling. We report an optimised protocol, enumerating the pitfalls encountered and precautions to be taken to maintain the robustness of the protocol. We characterise intermediate steps of small proteoliposome formation and the final formed GUVs. We show that the integrin molecules are successfully incorporated and are functional.
Collapse
Affiliation(s)
- Mariem Souissi
- Aix Marseille Univ, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINAM), Turing Centre for Living Systems, 13009 Marseille, France;
| | - Julien Pernier
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France; (J.P.); (C.L.C.)
| | - Olivier Rossier
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France; (O.R.); (G.G.)
| | - Gregory Giannone
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France; (O.R.); (G.G.)
| | - Christophe Le Clainche
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France; (J.P.); (C.L.C.)
| | - Emmanuèle Helfer
- Aix Marseille Univ, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINAM), Turing Centre for Living Systems, 13009 Marseille, France;
| | - Kheya Sengupta
- Aix Marseille Univ, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINAM), Turing Centre for Living Systems, 13009 Marseille, France;
| |
Collapse
|
28
|
Pettmann J, Huhn A, Abu Shah E, Kutuzov MA, Wilson DB, Dustin ML, Davis SJ, van der Merwe PA, Dushek O. The discriminatory power of the T cell receptor. eLife 2021; 10:e67092. [PMID: 34030769 PMCID: PMC8219380 DOI: 10.7554/elife.67092] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/15/2021] [Indexed: 12/20/2022] Open
Abstract
T cells use their T cell receptors (TCRs) to discriminate between lower-affinity self and higher-affinity non-self peptides presented on major histocompatibility complex (pMHC) antigens. Although the discriminatory power of the TCR is widely believed to be near-perfect, technical difficulties have hampered efforts to precisely quantify it. Here, we describe a method for measuring very low TCR/pMHC affinities and use it to measure the discriminatory power of the TCR and the factors affecting it. We find that TCR discrimination, although enhanced compared with conventional cell-surface receptors, is imperfect: primary human T cells can respond to pMHC with affinities as low as KD ∼ 1 mM. The kinetic proofreading mechanism fit our data, providing the first estimates of both the time delay (2.8 s) and number of biochemical steps (2.67) that are consistent with the extraordinary sensitivity of antigen recognition. Our findings explain why self pMHC frequently induce autoimmune diseases and anti-tumour responses, and suggest ways to modify TCR discrimination.
Collapse
Affiliation(s)
- Johannes Pettmann
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
- Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Anna Huhn
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| | - Enas Abu Shah
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
- Kennedy Institute of Rheumatology, University of OxfordOxfordUnited Kingdom
| | - Mikhail A Kutuzov
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| | - Daniel B Wilson
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
- Boston University, Department of Mathematics and StatisticsBostonUnited States
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of OxfordOxfordUnited Kingdom
| | - Simon J Davis
- Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | | | - Omer Dushek
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
29
|
Abstract
INTRODUCTION Proteomics, i.e. the study of the set of proteins produced in a cell, tissue, organism, or biological entity, has made possible analyses and contextual comparisons of proteomes/proteins and biological functions among the most disparate entities, from viruses to the human being. In this way, proteomic scrutiny of tumor-associated proteins, autoantigens, and pathogen antigens offers the tools for fighting cancer, autoimmunity, and infections. AREAS COVERED Comparative proteomics and immunoproteomics, the new scientific disciplines generated by proteomics, are the main themes of the present review that describes how comparative analyses of pathogen and human proteomes led to re-modulate the molecular mimicry concept of the pre-proteomic era. I.e. before proteomics, molecular mimicry - the sharing of peptide sequences between two biological entities - was considered as intrinsically endowed with immunologic properties and was related to cross-reactivity. Proteomics allowed to redefine such an assumption using physicochemical parameters according to which frequency and hydrophobicity preferentially confer an immunologic potential to shared peptide sequences. EXPERT OPINION Proteomics is outlining peptide platforms to be used for the diagnostics and management of human diseases. A Molecular Medicine targeted to obtain healing without paying the price for adverse events is on the horizon. The next step is to take up the challenge and operate the paradigm shift that the current proteomic era requires.
Collapse
Affiliation(s)
- Darja Kanduc
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
30
|
Lei Q, Guo J, Kong F, Cao J, Wang L, Zhu W, Brinker CJ. Bioinspired Cell Silicification: From Extracellular to Intracellular. J Am Chem Soc 2021; 143:6305-6322. [PMID: 33826324 DOI: 10.1021/jacs.1c00814] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In nature, biosilicification directs the formation of elaborate amorphous silica exoskeletons that provide diatoms mechanically strong, chemically inert, non-decomposable silica armor conferring chemical and thermal stability as well as resistance to microbial attack, without changing the optical transparency or adversely effecting nutrient and waste exchange required for growth. These extraordinary silica/cell biocomposites have inspired decades of biomimetic research aimed at replication of diatoms' hierarchically organized exoskeletons, immobilization of cells or living organisms within silica matrices and coatings to protect them against harmful external stresses, genetic re-programming of cellular functions by virtue of physico-chemical confinement within silica, cellular integration into devices, and endowment of cells with non-native, abiotic properties through facile silica functionalization. In this Perspective, we focus our discussions on the development and concomitant challenges of bioinspired cell silicification ranging from "cells encapsulated within 3D silica matrices" and "cells encapsulated within 2D silica shells" to extra- and intracellular silica replication, wherein all biomolecular interfaces are encased within nanoscopic layers of amorphous silica. We highlight notable examples of advances in the science and technology of biosilicification and consider challenges to advancing the field, where we propose cellular "mineralization" with arbitrary nanoparticle exoskeletons as a generalizable means to impart limitless abiotic properties and functions to cells, and, based on the interchangeability of water and silicic acid and analogies between amorphous ice and amorphous silica, we consider "freezing" cells within amorphous silica as an alternative to cryo-preservation.
Collapse
Affiliation(s)
- Qi Lei
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jimin Guo
- Center for Micro-Engineered Materials, Department of Chemical and Biological Engineering, The University of New Mexico, Albuquerque, New Mexico 87131, United States.,Department of Internal Medicine, Molecular Medicine, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Fanhui Kong
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jiangfan Cao
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Lu Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Wei Zhu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - C Jeffrey Brinker
- Center for Micro-Engineered Materials, Department of Chemical and Biological Engineering, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| |
Collapse
|
31
|
Yang X, Annaert W. The Nanoscopic Organization of Synapse Structures: A Common Basis for Cell Communication. MEMBRANES 2021; 11:248. [PMID: 33808285 PMCID: PMC8065904 DOI: 10.3390/membranes11040248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 11/16/2022]
Abstract
Synapse structures, including neuronal and immunological synapses, can be seen as the plasma membrane contact sites between two individual cells where information is transmitted from one cell to the other. The distance between the two plasma membranes is only a few tens of nanometers, but these areas are densely populated with functionally different proteins, including adhesion proteins, receptors, and transporters. The narrow space between the two plasma membranes has been a barrier for resolving the synaptic architecture due to the diffraction limit in conventional microscopy (~250 nm). Various advanced super-resolution microscopy techniques, such as stimulated emission depletion (STED), structured illumination microscopy (SIM), and single-molecule localization microscopy (SMLM), bypass the diffraction limit and provide a sub-diffraction-limit resolving power, ranging from 10 to 100 nm. The studies using super-resolution microscopy have revealed unprecedented details of the nanoscopic organization and dynamics of synaptic molecules. In general, most synaptic proteins appear to be heterogeneously distributed and form nanodomains at the membranes. These nanodomains are dynamic functional units, playing important roles in mediating signal transmission through synapses. Herein, we discuss our current knowledge on the super-resolution nanoscopic architecture of synapses and their functional implications, with a particular focus on the neuronal synapses and immune synapses.
Collapse
Affiliation(s)
| | - Wim Annaert
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Gasthuisberg, B-3000 Leuven, Belgium;
| |
Collapse
|
32
|
Ghosh S, Di Bartolo V, Tubul L, Shimoni E, Kartvelishvily E, Dadosh T, Feigelson SW, Alon R, Alcover A, Haran G. ERM-Dependent Assembly of T Cell Receptor Signaling and Co-stimulatory Molecules on Microvilli prior to Activation. Cell Rep 2021; 30:3434-3447.e6. [PMID: 32160548 DOI: 10.1016/j.celrep.2020.02.069] [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: 08/27/2019] [Revised: 12/16/2019] [Accepted: 02/18/2020] [Indexed: 01/25/2023] Open
Abstract
T cell surfaces are covered with microvilli, actin-rich and flexible protrusions. We use super-resolution microscopy to show that ≥90% of T cell receptor (TCR) complex molecules TCRαβ and TCRζ, as well as the co-receptor CD4 (cluster of differentiation 4) and the co-stimulatory molecule CD2, reside on microvilli of resting human T cells. Furthermore, TCR proximal signaling molecules involved in the initial stages of the immune response, including the protein tyrosine kinase Lck (lymphocyte-specific protein tyrosine kinase) and the key adaptor LAT (linker for activation of T cells), are also enriched on microvilli. Notably, phosphorylated proteins of the ERM (ezrin, radixin, and moesin) family colocalize with TCRαβ as well as with actin filaments, implying a role for one or more ERMs in linking the TCR complex to the actin cytoskeleton within microvilli. Our results establish microvilli as key signaling hubs, in which the TCR complex and its proximal signaling molecules and adaptors are preassembled prior to activation in an ERM-dependent manner, facilitating initial antigen sensing.
Collapse
Affiliation(s)
- Shirsendu Ghosh
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France
| | - Liron Tubul
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eyal Shimoni
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elena Kartvelishvily
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tali Dadosh
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sara W Feigelson
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Andres Alcover
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France
| | - Gilad Haran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
33
|
Dam T, Junghans V, Humphrey J, Chouliara M, Jönsson P. Calcium Signaling in T Cells Is Induced by Binding to Nickel-Chelating Lipids in Supported Lipid Bilayers. Front Physiol 2021; 11:613367. [PMID: 33551841 PMCID: PMC7859345 DOI: 10.3389/fphys.2020.613367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/30/2020] [Indexed: 12/26/2022] Open
Abstract
Supported lipid bilayers (SLBs) are one of the most common cell-membrane model systems to study cell-cell interactions. Nickel-chelating lipids are frequently used to functionalize the SLB with polyhistidine-tagged ligands. We show here that these lipids by themselves can induce calcium signaling in T cells, also when having protein ligands on the SLB. This is important to avoid "false" signaling events in cell studies with SLBs, but also to better understand the molecular mechanisms involved in T-cell signaling. Jurkat T cells transfected with the non-signaling molecule rat CD48 were found to bind to ligand-free SLBs containing ≥2 wt% nickel-chelating lipids upon which calcium signaling was induced. This signaling fraction steadily increased from 24 to 60% when increasing the amount of nickel-chelating lipids from 2 to 10 wt%. Both the signaling fraction and signaling time did not change significantly compared to ligand-free SLBs when adding the CD48-ligand rat CD2 to the SLB. Blocking the SLB with bovine serum albumin reduced the signaling fraction to 11%, while preserving CD2 binding and the exclusion of the phosphatase CD45 from the cell-SLB contacts. Thus, CD45 exclusion alone was not sufficient to result in calcium signaling. In addition, more cells signaled on ligand-free SLBs with copper-chelating lipids instead of nickel-chelating lipids and the signaling was found to be predominantly via T-cell receptor (TCR) triggering. Hence, it is possible that the nickel-chelating lipids act as ligands to the cell's TCRs, an interaction that needs to be blocked to avoid unwanted cell activation.
Collapse
Affiliation(s)
- Tommy Dam
- Department of Chemistry, Lund University, Lund, Sweden
| | | | - Jane Humphrey
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | | | - Peter Jönsson
- Department of Chemistry, Lund University, Lund, Sweden
| |
Collapse
|
34
|
Zhang W, Yang Y, Cui B. New perspectives on the roles of nanoscale surface topography in modulating intracellular signaling. CURRENT OPINION IN SOLID STATE & MATERIALS SCIENCE 2021; 25:100873. [PMID: 33364912 PMCID: PMC7751896 DOI: 10.1016/j.cossms.2020.100873] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The physical properties of biomaterials, such as elasticity, stiffness, and surface nanotopography, are mechanical cues that regulate a broad spectrum of cell behaviors, including migration, differentiation, proliferation, and reprogramming. Among them, nanoscale surface topography, i.e. nanotopography, defines the nanoscale shape and spatial arrangement of surface elements, which directly interact with the cell membranes and stimulate changes in the cell signaling pathways. In biological systems, the effects of nanotopography are often entangled with those of other mechanical and biochemical factors. Precise engineering of 2D nanopatterns and 3D nanostructures with well-defined features has provided a powerful means to study the cellular responses to specific topographic features. In this Review, we discuss efforts in the last three years to understand how nanotopography affects membrane receptor activation, curvature-induced cell signaling, and stem cell differentiation.
Collapse
Affiliation(s)
| | | | - Bianxiao Cui
- Department of Chemistry, Stanford University, ChEM-H/Wu Tsai Neuroscience Research Complex, S285, 290 Jane Stanford Way, Stanford, CA, 94305, United States
| |
Collapse
|
35
|
Aramesh M, Mergenthal S, Issler M, Plochberger B, Weber F, Qin XH, Liska R, Duda GN, Huppa JB, Ries J, Schütz GJ, Klotzsch E. Functionalized Bead Assay to Measure Three-dimensional Traction Forces during T-cell Activation. NANO LETTERS 2021; 21:507-514. [PMID: 33305952 DOI: 10.1021/acs.nanolett.0c03964] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
When T-cells probe their environment for antigens, the bond between the T-cell receptor (TCR) and the peptide-loaded major histocompatibility complex (MHC) is put under tension, thereby influencing the antigen discrimination. Yet, the quantification of such forces in the context of T-cell signaling is technically challenging. Here, we developed a traction force microscopy platform which allows for quantifying the pulls and pushes exerted via T-cell microvilli, in both tangential and normal directions, during T-cell activation. We immobilized specific T-cell activating antibodies on the marker beads used to read out the hydrogel deformation. Microvilli targeted the functionalized beads, as confirmed by superresolution microscopy of the local actin organization. Moreover, we found that cellular components, such as actin, TCR, and CD45 reorganize upon interaction with the beads, such that actin forms a vortex-like ring structure around the beads and TCR is enriched at the bead surface, whereas CD45 is excluded from bead-microvilli contacts.
Collapse
Affiliation(s)
- Morteza Aramesh
- Laboratory of Applied Mechanobiology, Department for Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Simon Mergenthal
- Institute of Biology, Experimental Biophysics/Mechanobiology, Humboldt Universität zu Berlin, 10115 Berlin, Germany
| | - Marcel Issler
- Institute of Biology, Experimental Biophysics/Mechanobiology, Humboldt Universität zu Berlin, 10115 Berlin, Germany
| | - Birgit Plochberger
- Upper Austria University of Applied Sciences, Campus Linz, Garnisonstrasse 21, 4020 Linz, Austria
| | - Florian Weber
- Upper Austria University of Applied Sciences, Campus Linz, Garnisonstrasse 21, 4020 Linz, Austria
| | - Xiao-Hua Qin
- Institute for Biomechanics, Department for Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Robert Liska
- Institute of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9/163/MC, 1060 Vienna, Austria
| | - Georg N Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin, BIH Center for Regenerative Therapies, Berlin Institute of Health, 13353 Berlin, Germany
| | - Johannes B Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Jonas Ries
- EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | | | - Enrico Klotzsch
- Laboratory of Applied Mechanobiology, Department for Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
- Institute of Biology, Experimental Biophysics/Mechanobiology, Humboldt Universität zu Berlin, 10115 Berlin, Germany
| |
Collapse
|
36
|
Lippert AH, Dimov IB, Winkel AK, Humphrey J, McColl J, Chen KY, Santos AM, Jenkins E, Franze K, Davis SJ, Klenerman D. Soft Polydimethylsiloxane-Supported Lipid Bilayers for Studying T Cell Interactions. Biophys J 2021; 120:35-45. [PMID: 33248128 PMCID: PMC7820804 DOI: 10.1016/j.bpj.2020.11.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/22/2020] [Accepted: 11/17/2020] [Indexed: 12/23/2022] Open
Abstract
Much of what we know about the early stages of T cell activation has been obtained from studies of T cells interacting with glass-supported lipid bilayers that favor imaging but are orders of magnitude stiffer than typical cells. We developed a method for attaching lipid bilayers to polydimethylsiloxane polymer supports, producing "soft bilayers" with physiological levels of mechanical resistance (Young's modulus of 4 kPa). Comparisons of T cell behavior on soft and glass-supported bilayers revealed that whereas late stages of T cell activation are thought to be substrate-stiffness dependent, early calcium signaling was unaffected by substrate rigidity, implying that early steps in T cell receptor triggering are not mechanosensitive. The exclusion of large receptor-type phosphatases was observed on the soft bilayers, however, even though it is yet to be demonstrated at authentic cell-cell contacts. This work sets the stage for an imaging-based exploration of receptor signaling under conditions closely mimicking physiological cell-cell contact.
Collapse
Affiliation(s)
- Anna H Lippert
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom.
| | - Ivan B Dimov
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Alexander K Winkel
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Jane Humphrey
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - James McColl
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Kevin Y Chen
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Ana M Santos
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Edward Jenkins
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Simon J Davis
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
37
|
Li R, Ma C, Cai H, Chen W. The CAR T-Cell Mechanoimmunology at a Glance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002628. [PMID: 33344135 PMCID: PMC7740088 DOI: 10.1002/advs.202002628] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/13/2020] [Indexed: 05/10/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell transfer is a novel paradigm of adoptive T-cell immunotherapy. When coming into contact with a target cancer cell, CAR T-cell forms a nonclassical immunological synapse with the cancer cell and dynamically orchestrates multiple critical forces to commit cytotoxic immune function. Such an immunologic process involves a force transmission in the CAR and a spatiotemporal remodeling of cell cytoskeleton to facilitate CAR activation and CAR T-cell cytotoxic function. Yet, the detailed understanding of such mechanotransduction at the interface between the CAR T-cell and the target cell, as well as its molecular structure and signaling, remains less defined and is just beginning to emerge. This article summarizes the basic mechanisms and principles of CAR T-cell mechanoimmunology, and various lessons that can be comparatively learned from interrogation of mechanotransduction at the immunological synapse in normal cytotoxic T-cell. The recent development and future application of novel bioengineering tools for studying CAR T-cell mechanoimmunology is also discussed. It is believed that this progress report will shed light on the CAR T-cell mechanoimmunology and encourage future researches in revealing the less explored yet important mechanosensing and mechanotransductive mechanisms involved in CAR T-cell immuno-oncology.
Collapse
Affiliation(s)
- Rui Li
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNY11201USA
- Department of Biomedical EngineeringNew York UniversityBrooklynNY11201USA
| | - Chao Ma
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNY11201USA
- Department of Biomedical EngineeringNew York UniversityBrooklynNY11201USA
| | - Haogang Cai
- Tech4Health instituteNYU Langone HealthNew YorkNY10016USA
- Department of RadiologyNYU Langone HealthNew YorkNY10016USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNY11201USA
- Department of Biomedical EngineeringNew York UniversityBrooklynNY11201USA
- Laura and Isaac Perlmutter Cancer CenterNYU Langone HealthNew YorkNY10016USA
| |
Collapse
|
38
|
Rukhlenko O, Kholodenko BN, Kolch W. Systems biology approaches to macromolecules: the role of dynamic protein assemblies in information processing. Curr Opin Struct Biol 2020; 67:61-68. [PMID: 33126139 DOI: 10.1016/j.sbi.2020.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/10/2020] [Accepted: 09/16/2020] [Indexed: 11/17/2022]
Abstract
Macromolecular protein assemblies govern many cellular processes and are disturbed in many diseases including cancer. Often seen as static molecular machines, protein complexes involved in signal transduction networks exhibit intricate dynamics that are critical for their function. Using the RAS-RAF-MEK-ERK pathway as example we discuss recent progress in our understanding of protein complex dynamics achieved through mathematical modelling, computational simulations and structural studies. The emerging picture highlights that both spatial and temporal dynamics cooperate to enable correct signal processing and the fine tuning of timing, duration and strengths of signalling. These dynamic processes are subverted by oncogenic mutations and contribute to tumorigenesis and drug resistance.
Collapse
Affiliation(s)
- Oleksii Rukhlenko
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Boris N Kholodenko
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
39
|
Lee CH, Salio M, Napolitani G, Ogg G, Simmons A, Koohy H. Predicting Cross-Reactivity and Antigen Specificity of T Cell Receptors. Front Immunol 2020; 11:565096. [PMID: 33193332 PMCID: PMC7642207 DOI: 10.3389/fimmu.2020.565096] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
Adaptive immune recognition is mediated by specific interactions between heterodimeric T cell receptors (TCRs) and their cognate peptide-MHC (pMHC) ligands, and the methods to accurately predict TCR:pMHC interaction would have profound clinical, therapeutic and pharmaceutical applications. Herein, we review recent developments in predicting cross-reactivity and antigen specificity of TCR recognition. We discuss current experimental and computational approaches to investigate cross-reactivity and antigen-specificity of TCRs and highlight how integrating kinetic, biophysical and structural features may offer valuable insights in modeling immunogenicity. We further underscore the close inter-relationship of these two interconnected notions and the need to investigate each in the light of the other for a better understanding of T cell responsiveness for the effective clinical applications.
Collapse
Affiliation(s)
- Chloe H. Lee
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Mariolina Salio
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Giorgio Napolitani
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Graham Ogg
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Alison Simmons
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford, United Kingdom
| | - Hashem Koohy
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
40
|
Mørch AM, Bálint Š, Santos AM, Davis SJ, Dustin ML. Coreceptors and TCR Signaling - the Strong and the Weak of It. Front Cell Dev Biol 2020; 8:597627. [PMID: 33178706 PMCID: PMC7596257 DOI: 10.3389/fcell.2020.597627] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/28/2020] [Indexed: 12/02/2022] Open
Abstract
The T-cell coreceptors CD4 and CD8 have well-characterized and essential roles in thymic development, but how they contribute to immune responses in the periphery is unclear. Coreceptors strengthen T-cell responses by many orders of magnitude - beyond a million-fold according to some estimates - but the mechanisms underlying these effects are still debated. T-cell receptor (TCR) triggering is initiated by the binding of the TCR to peptide-loaded major histocompatibility complex (pMHC) molecules on the surfaces of other cells. CD4 and CD8 are the only T-cell proteins that bind to the same pMHC ligand as the TCR, and can directly associate with the TCR-phosphorylating kinase Lck. At least three mechanisms have been proposed to explain how coreceptors so profoundly amplify TCR signaling: (1) the Lck recruitment model and (2) the pseudodimer model, both invoked to explain receptor triggering per se, and (3) two-step coreceptor recruitment to partially triggered TCRs leading to signal amplification. More recently it has been suggested that, in addition to initiating or augmenting TCR signaling, coreceptors effect antigen discrimination. But how can any of this be reconciled with TCR signaling occurring in the absence of CD4 or CD8, and with their interactions with pMHC being among the weakest specific protein-protein interactions ever described? Here, we review each theory of coreceptor function in light of the latest structural, biochemical, and functional data. We conclude that the oldest ideas are probably still the best, i.e., that their weak binding to MHC proteins and efficient association with Lck allow coreceptors to amplify weak incipient triggering of the TCR, without comprising TCR specificity.
Collapse
Affiliation(s)
- Alexander M. Mørch
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Štefan Bálint
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Simon J. Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Michael L. Dustin
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
41
|
Orbach R, Su X. Surfing on Membrane Waves: Microvilli, Curved Membranes, and Immune Signaling. Front Immunol 2020; 11:2187. [PMID: 33013920 PMCID: PMC7516127 DOI: 10.3389/fimmu.2020.02187] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/11/2020] [Indexed: 01/22/2023] Open
Abstract
Microvilli are finger-like membrane protrusions, supported by the actin cytoskeleton, and found on almost all cell types. A growing body of evidence suggests that the dynamic lymphocyte microvilli, with their highly curved membranes, play an important role in signal transduction leading to immune responses. Nevertheless, challenges in modulating local membrane curvature and monitoring the high dynamicity of microvilli hampered the investigation of the curvature-generation mechanism and its functional consequences in signaling. These technical barriers have been partially overcome by recent advancements in adapted super-resolution microscopy. Here, we review the up-to-date progress in understanding the mechanisms and functional consequences of microvillus formation in T cell signaling. We discuss how the deformation of local membranes could potentially affect the organization of signaling proteins and their biochemical activities. We propose that curved membranes, together with the underlying cytoskeleton, shape microvilli into a unique compartment that sense and process signals leading to lymphocyte activation.
Collapse
Affiliation(s)
- Ron Orbach
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, United States
| | - Xiaolei Su
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, United States
- Yale Cancer Center, Yale University, New Haven, CT, United States
| |
Collapse
|
42
|
Farrell MV, Webster S, Gaus K, Goyette J. T Cell Membrane Heterogeneity Aids Antigen Recognition and T Cell Activation. Front Cell Dev Biol 2020; 8:609. [PMID: 32850786 PMCID: PMC7399036 DOI: 10.3389/fcell.2020.00609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022] Open
Abstract
T cells are critical for co-ordinating the immune response. T cells are activated when their surface T cell receptors (TCRs) engage cognate antigens in the form of peptide-major histocompatibility complexes (pMHC) presented on the surface of antigen presenting cells (APCs). Large changes in the contact interface between T cells and APCs occur over the course of tens of minutes from the initial contact to the formation of a large-scale junction between the two cells. The mature junction between a T cell and APC is known as the immunological synapse, and this specialized plasma membrane structure is the major platform for TCR signaling. It has long been known that the complex organization of signaling molecules at the synapse is critical for appropriate activation of T cells, but within the last decade advances in microscopy have opened up investigation into the dynamics of T cell surface topology in the immune synapse. From mechanisms mediating the initial contact between T cells and APCs to roles in the organization of molecules in the mature synapse, these studies have made it increasingly clear that local membrane topology has a large impact on signaling processes. This review focuses on the functional consequences of the T cells' highly dynamic and heterogeneous membrane, in particular, how membrane topology leads to the reorganization of membrane proteins on the T cell surface.
Collapse
Affiliation(s)
- Megan V Farrell
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Samantha Webster
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
43
|
Teimouri H, Kolomeisky AB. Relaxation Times of Ligand-Receptor Complex Formation Control T Cell Activation. Biophys J 2020; 119:182-189. [PMID: 32562619 DOI: 10.1016/j.bpj.2020.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/20/2020] [Accepted: 06/01/2020] [Indexed: 11/18/2022] Open
Abstract
One of the most important functions of immune T cells is to recognize the presence of the pathogen-derived ligands and to quickly respond to them while at the same time not responding to its own ligands. This is known as absolute discrimination, and it is one of the most challenging phenomena to explain. The effectiveness of pathogen detection by T cell receptor is limited by chemical similarity of foreign and self-peptides and very low concentrations of foreign ligands. We propose a new mechanism of how absolute discrimination by T cells might function. It is suggested that the decision to activate or not to activate the immune response is controlled by the time to reach the stationary concentration of the T-cell-receptor-ligand-activated complex, which transfers the signal to downstream cellular biochemical networks. Our theoretical method models T cell receptor phosphorylation events as a sequence of stochastic transitions between discrete biochemical states, and this allows us to explicitly describe the dynamical properties of the system. It is found that the proposed criterion on the relaxation times is able to explain available experimental observations. In addition, we suggest that the level of stochastic noise might be an additional factor in the activation mechanisms. Furthermore, our theoretical approach explicitly analyzes the relationships between speed, sensitivity, and specificity of T cell functioning, which are the main characteristics of the process. Thus, it clarifies the molecular picture of T cell activation in immune response.
Collapse
Affiliation(s)
- Hamid Teimouri
- Department of Chemistry, Rice University, Houston, Texas; Center for Theoretical Biological Physics, Rice University, Houston, Texas
| | - Anatoly B Kolomeisky
- Department of Chemistry, Rice University, Houston, Texas; Center for Theoretical Biological Physics, Rice University, Houston, Texas; Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas; Department of Physics and Astronomy, Rice University, Houston, Texas.
| |
Collapse
|
44
|
Zhao LP, Papadopoulos GK, Kwok WW, Moustakas AK, Bondinas GP, Larsson HE, Ludvigsson J, Marcus C, Samuelsson U, Wang R, Pyo CW, Nelson WC, Geraghty DE, Lernmark Å. Motifs of Three HLA-DQ Amino Acid Residues (α44, β57, β135) Capture Full Association With the Risk of Type 1 Diabetes in DQ2 and DQ8 Children. Diabetes 2020; 69:1573-1587. [PMID: 32245799 PMCID: PMC7306123 DOI: 10.2337/db20-0075] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/30/2020] [Indexed: 12/25/2022]
Abstract
HLA-DQA1 and -DQB1 are strongly associated with type 1 diabetes (T1D), and DQ8.1 and DQ2.5 are major risk haplotypes. Next-generation targeted sequencing of HLA-DQA1 and -DQB1 in Swedish newly diagnosed 1- to 18 year-old patients (n = 962) and control subjects (n = 636) was used to construct abbreviated DQ haplotypes, converted into amino acid (AA) residues, and assessed for their associations with T1D. A hierarchically organized haplotype (HOH) association analysis allowed 45 unique DQ haplotypes to be categorized into seven clusters. The DQ8/9 cluster included two DQ8.1 risk and the DQ9 resistant haplotypes, and the DQ2 cluster included the DQ2.5 risk and DQ2.2 resistant haplotypes. Within each cluster, HOH found residues α44Q (odds ratio [OR] 3.29, P = 2.38 * 10-85) and β57A (OR 3.44, P = 3.80 * 10-84) to be associated with T1D in the DQ8/9 cluster representing all ten residues (α22, α23, α44, α49, α51, α53, α54, α73, α184, β57) due to complete linkage disequilibrium (LD) of α44 with eight such residues. Within the DQ2 cluster and due to LD, HOH analysis found α44C and β135D to share the risk for T1D (OR 2.10, P = 1.96 * 10-20). The motif "QAD" of α44, β57, and β135 captured the T1D risk association of DQ8.1 (OR 3.44, P = 3.80 * 10-84), and the corresponding motif "CAD" captured the risk association of DQ2.5 (OR 2.10, P = 1.96 * 10-20). Two risk associations were related to GAD65 autoantibody (GADA) and IA-2 autoantibody (IA-2A) but in opposite directions. CAD was positively associated with GADA (OR 1.56, P = 6.35 * 10-8) but negatively with IA-2A (OR 0.59, P = 6.55 * 10-11). QAD was negatively associated with GADA (OR 0.88; P = 3.70 * 10-3) but positively with IA-2A (OR 1.64; P = 2.40 * 10-14), despite a single difference at α44. The residues are found in and around anchor pockets 1 and 9, as potential T-cell receptor contacts, in the areas for CD4 binding and putative homodimer formation. The identification of three HLA-DQ AAs (α44, β57, β135) conferring T1D risk should sharpen functional and translational studies.
Collapse
Affiliation(s)
- Lue Ping Zhao
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - George K Papadopoulos
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Antonis K Moustakas
- Department of Food Science and Technology, Faculty of Environmental Sciences, Ionian University, Argostoli, Cephalonia, Greece
| | - George P Bondinas
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - Helena Elding Larsson
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital, Region Östergötland, and Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Claude Marcus
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Samuelsson
- Crown Princess Victoria Children's Hospital, Region Östergötland, and Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Ruihan Wang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Chul-Woo Pyo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Wyatt C Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Daniel E Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
45
|
Hilzenrat G, Pandžić E, Yang Z, Nieves DJ, Goyette J, Rossy J, Ma Y, Gaus K. Conformational States Control Lck Switching between Free and Confined Diffusion Modes in T Cells. Biophys J 2020; 118:1489-1501. [PMID: 32097620 PMCID: PMC7091564 DOI: 10.1016/j.bpj.2020.01.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 11/13/2022] Open
Abstract
T cell receptor phosphorylation by Lck is an essential step in T cell activation. It is known that the conformational states of Lck control enzymatic activity; however, the underlying principles of how Lck finds its substrate over the plasma membrane remain elusive. Here, single-particle tracking is paired with photoactivatable localization microscopy to observe the diffusive modes of Lck in the plasma membrane. Individual Lck molecules switched between free and confined diffusion in both resting and stimulated T cells. Lck mutants locked in the open conformation were more confined than Lck mutants in the closed conformation. Further confinement of kinase-dead versions of Lck suggests that Lck confinement was not caused by phosphorylated substrates. Our data support a model in which confined diffusion of open Lck results in high local phosphorylation rates, and inactive, closed Lck diffuses freely to enable long-range distribution over the plasma membrane.
Collapse
Affiliation(s)
- Geva Hilzenrat
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia; Commonwealth Scientific and Industry Research Organization (CSIRO), Manufacturing, Clayton, Victoria, Australia
| | - Elvis Pandžić
- BioMedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, Australia
| | - Zhengmin Yang
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia
| | - Daniel J Nieves
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia; Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia
| | - Jérémie Rossy
- Biotechnology Institute Thurgau, University of Konstanz, Kreuzlingen, Switzerland
| | - Yuanqing Ma
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia.
| |
Collapse
|
46
|
Rushdi M, Li K, Yuan Z, Travaglino S, Grakoui A, Zhu C. Mechanotransduction in T Cell Development, Differentiation and Function. Cells 2020; 9:E364. [PMID: 32033255 PMCID: PMC7072571 DOI: 10.3390/cells9020364] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 02/07/2023] Open
Abstract
Cells in the body are actively engaging with their environments that include both biochemical and biophysical aspects. The process by which cells convert mechanical stimuli from their environment to intracellular biochemical signals is known as mechanotransduction. Exemplifying the reliance on mechanotransduction for their development, differentiation and function are T cells, which are central to adaptive immune responses. T cell mechanoimmunology is an emerging field that studies how T cells sense, respond and adapt to the mechanical cues that they encounter throughout their life cycle. Here we review different stages of the T cell's life cycle where existing studies have shown important effects of mechanical force or matrix stiffness on a T cell as sensed through its surface molecules, including modulating receptor-ligand interactions, inducing protein conformational changes, triggering signal transduction, amplifying antigen discrimination and ensuring directed targeted cell killing. We suggest that including mechanical considerations in the immunological studies of T cells would inform a more holistic understanding of their development, differentiation and function.
Collapse
Affiliation(s)
- Muaz Rushdi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.R.); (K.L.); (S.T.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Kaitao Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.R.); (K.L.); (S.T.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Zhou Yuan
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30313, USA
| | - Stefano Travaglino
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.R.); (K.L.); (S.T.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Arash Grakoui
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, GA 30329, USA;
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.R.); (K.L.); (S.T.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30313, USA
| |
Collapse
|
47
|
Núñez MF, Wisser K, Veatch SL. Synergistic factors control kinase-phosphatase organization in B-cells engaged with supported bilayers. Mol Biol Cell 2019; 31:667-682. [PMID: 31877064 PMCID: PMC7202075 DOI: 10.1091/mbc.e19-09-0507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
B-cells become activated by ligands with varying valency and mode of presentation to the B-cell receptor (BCR). We previously demonstrated that clustering the immunoglobulin M (IgM) isotype of BCR with an artificial soluble cross-linker stabilized an ordered phase-like domain that enriched kinases and depleted phosphatases to promote receptor tyrosine phosphorylation. BCR is also activated by ligands presented at surfaces, and here we activate B-cells via supported bilayers of phosphatidylcholine lipids, a natural ligand for the IgM BCR expressed in the CH27 cells used. Using superresolution fluorescence localization microscopy, along with a quantitative cross-correlation analysis, we find that BRCs engaged with bilayers sort minimal peptide markers of liquid-ordered and liquid-disordered phases, indicating that ordered-domain stabilization is a general feature of BCR clustering. The phosphatase CD45 is more strongly excluded from bilayer-engaged BRCs than a transmembrane peptide, indicating that mechanisms other than domain partitioning contribute to its organization. Experimental observations are assembled into a minimal model of receptor activation that incorporates both ordered domains and direct phosphatase exclusion mechanisms to produce a more sensitive response.
Collapse
Affiliation(s)
| | - Kathleen Wisser
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48105
| | - Sarah L Veatch
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48105
| |
Collapse
|
48
|
Hoober JK, Eggink LL, Cote R. Stories From the Dendritic Cell Guardhouse. Front Immunol 2019; 10:2880. [PMID: 31921144 PMCID: PMC6919295 DOI: 10.3389/fimmu.2019.02880] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/25/2019] [Indexed: 12/19/2022] Open
Abstract
Phagocytic cells [dendritic cells (DCs), macrophages, monocytes, neutrophils, and mast cells] utilize C-type (Ca2+-dependent) lectin-like (CLEC) receptors to identify and internalize pathogens or danger signals. As monitors of environmental imbalances, CLEC receptors are particularly important in the function of DCs. Activation of the immune system requires, in sequence, presentation of antigen to the T cell receptor (TCR) by DCs, interaction of co-stimulatory factors such as CD40/80/86 on DCs with CD40L and CD28 on T cells, and production of IL-12 and/or IFN-α/β to amplify T cell differentiation and expansion. Without this sequence of events within an inflammatory environment, or in a different order, antigen-specific T cells become unresponsive, are deleted or become regulatory T cells. Thus, the mode by which CLEC receptors on DCs are engaged can either elicit activation of T cells to achieve an immune response or induce tolerance. This minireview illustrates these aspects with Dectin-1, DEC205, the mannose receptor and CLEC10A as examples.
Collapse
Affiliation(s)
| | | | - Robert Cote
- Susavion Biosciences, Inc., Tempe, AZ, United States
| |
Collapse
|
49
|
Kaitao L, William R, Zhou Y, Cheng Z. Single-molecule investigations of T-cell activation. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019; 12:102-110. [PMID: 32296738 PMCID: PMC7158867 DOI: 10.1016/j.cobme.2019.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
T-cell activation is the central event governing its development, differentiation, and effector functions. T-cell activation is initiated by the direct physical interaction of the T cell antigen receptor (TCR) with cognate peptide presented by the major histocompatibility complex (pMHC) molecule expressed on the antigen presenting cell (APC) surface. Since the identification of TCR as the only receptor for antigen on T cells three decades ago, studies have elucidated the major molecular players and signaling events responding to TCR stimulation. However, the question of how the physical event of pMHC binding is converted across the membrane into chemical events to initiate signal transduction remains elusive. Here we review recent investigations of T-cell activation using single-molecule force and fluorescence techniques that shed new light on this key question.
Collapse
Affiliation(s)
- Li Kaitao
- Wallace H. Coulter Department of Biomedical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Rittase William
- Wallace H. Coulter Department of Biomedical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Yuan Zhou
- George W. Woodruff School of Mechanical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Zhu Cheng
- Wallace H. Coulter Department of Biomedical Engineering
- George W. Woodruff School of Mechanical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|