1
|
Hu Y, Rogers J, Duan Y, Velusamy A, Narum S, Al Abdullatif S, Salaita K. Quantifying T cell receptor mechanics at membrane junctions using DNA origami tension sensors. NATURE NANOTECHNOLOGY 2024; 19:1674-1685. [PMID: 39103452 DOI: 10.1038/s41565-024-01723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 06/21/2024] [Indexed: 08/07/2024]
Abstract
The T cell receptor (TCR) is thought to be a mechanosensor, meaning that it transmits mechanical force to its antigen and leverages the force to amplify the specificity and magnitude of TCR signalling. Although a variety of molecular probes have been proposed to quantify TCR mechanics, these probes are immobilized on hard substrates, and thus fail to reveal fluid TCR-antigen interactions in the physiological context of cell membranes. Here we developed DNA origami tension sensors (DOTS) which bear force sensors on a DNA origami breadboard and allow mapping of TCR mechanotransduction at dynamic intermembrane junctions. We quantified the mechanical forces at fluid TCR-antigen bonds and observed their dependence on cell state, antigen mobility, antigen potency, antigen height and F-actin activity. The programmability of DOTS allows us to tether these to microparticles to mechanically screen antigens in high throughput using flow cytometry. Additionally, DOTS were anchored onto live B cells, allowing quantification of TCR mechanics at immune cell-cell junctions.
Collapse
Affiliation(s)
- Yuesong Hu
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Jhordan Rogers
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Yuxin Duan
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | | | - Steven Narum
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Rogers J, Bajur AT, Salaita K, Spillane KM. Mechanical control of antigen detection and discrimination by T and B cell receptors. Biophys J 2024; 123:2234-2255. [PMID: 38794795 PMCID: PMC11331051 DOI: 10.1016/j.bpj.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
The adaptive immune response is orchestrated by just two cell types, T cells and B cells. Both cells possess the remarkable ability to recognize virtually any antigen through their respective antigen receptors-the T cell receptor (TCR) and B cell receptor (BCR). Despite extensive investigations into the biochemical signaling events triggered by antigen recognition in these cells, our ability to predict or control the outcome of T and B cell activation remains elusive. This challenge is compounded by the sensitivity of T and B cells to the biophysical properties of antigens and the cells presenting them-a phenomenon we are just beginning to understand. Recent insights underscore the central role of mechanical forces in this process, governing the conformation, signaling activity, and spatial organization of TCRs and BCRs within the cell membrane, ultimately eliciting distinct cellular responses. Traditionally, T cells and B cells have been studied independently, with researchers working in parallel to decipher the mechanisms of activation. While these investigations have unveiled many overlaps in how these cell types sense and respond to antigens, notable differences exist. To fully grasp their biology and harness it for therapeutic purposes, these distinctions must be considered. This review compares and contrasts the TCR and BCR, placing emphasis on the role of mechanical force in regulating the activity of both receptors to shape cellular and humoral adaptive immune responses.
Collapse
Affiliation(s)
- Jhordan Rogers
- Department of Chemistry, Emory University, Atlanta, Georgia
| | - Anna T Bajur
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.
| | - Katelyn M Spillane
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom; Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
3
|
Jeffreys N, Brockman JM, Zhai Y, Ingber DE, Mooney DJ. Mechanical forces amplify TCR mechanotransduction in T cell activation and function. APPLIED PHYSICS REVIEWS 2024; 11:011304. [PMID: 38434676 PMCID: PMC10848667 DOI: 10.1063/5.0166848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/08/2023] [Indexed: 03/05/2024]
Abstract
Adoptive T cell immunotherapies, including engineered T cell receptor (eTCR) and chimeric antigen receptor (CAR) T cell immunotherapies, have shown efficacy in treating a subset of hematologic malignancies, exhibit promise in solid tumors, and have many other potential applications, such as in fibrosis, autoimmunity, and regenerative medicine. While immunoengineering has focused on designing biomaterials to present biochemical cues to manipulate T cells ex vivo and in vivo, mechanical cues that regulate their biology have been largely underappreciated. This review highlights the contributions of mechanical force to several receptor-ligand interactions critical to T cell function, with central focus on the TCR-peptide-loaded major histocompatibility complex (pMHC). We then emphasize the role of mechanical forces in (i) allosteric strengthening of the TCR-pMHC interaction in amplifying ligand discrimination during T cell antigen recognition prior to activation and (ii) T cell interactions with the extracellular matrix. We then describe approaches to design eTCRs, CARs, and biomaterials to exploit TCR mechanosensitivity in order to potentiate T cell manufacturing and function in adoptive T cell immunotherapy.
Collapse
Affiliation(s)
| | | | - Yunhao Zhai
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
4
|
Suarez Rodriguez F, Sanlidag S, Sahlgren C. Mechanical regulation of the Notch signaling pathway. Curr Opin Cell Biol 2023; 85:102244. [PMID: 37783031 DOI: 10.1016/j.ceb.2023.102244] [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: 05/19/2023] [Revised: 08/13/2023] [Accepted: 09/03/2023] [Indexed: 10/04/2023]
Abstract
The mechanical regulation of Notch signaling is an emerging area of interest in cell biology. Notch is essential in many physiological processes in which mechanical stress plays an important role. This review provides an overview of the mechanoregulation of Notch signaling in multiple steps of the pathway. First, we discuss the current knowledge on the direct mechanoregulation of Notch receptor maturation and localization to the membrane and the effect of mechanical stress on the Notch components. Next, we explore how ligand-receptor interactions and membrane dynamics are possible subjects to mechano-regulation, emphasizing the role of cytoskeletal interactions, membrane stiffness, and endocytic complex formation. We further delve into the necessity of tension generation for negative regulatory region (NRR) domain unfolding, facilitated by ligand endocytosis and other microforces. Additionally, we examine the indirect mechano-regulation of S2 and S3 cleavages. Finally, we discuss the mechanoregulation of the Notch intracellular domain (NICD) trafficking and nuclear entry and the impact of mechanical stress on heterochromatin dynamics and nuclear NICD interactions. This review aims to draw attention to the intricate interplay between mechanical cues and Notch signaling regulation, offering novel insights into the multifaceted nature of cellular mechanobiology.
Collapse
Affiliation(s)
- Freddy Suarez Rodriguez
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, FI-20520, Turku, Finland; Turku Bioscience, Åbo Akademi University and University of Turku, Tykistökatu 6, FI-20520, Turku, Finland; InFLAMES Research Flagship Center, Åbo Akademi University and University of Turku, Turku, Finland
| | - Sami Sanlidag
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, FI-20520, Turku, Finland; Turku Bioscience, Åbo Akademi University and University of Turku, Tykistökatu 6, FI-20520, Turku, Finland; InFLAMES Research Flagship Center, Åbo Akademi University and University of Turku, Turku, Finland
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, FI-20520, Turku, Finland; Turku Bioscience, Åbo Akademi University and University of Turku, Tykistökatu 6, FI-20520, Turku, Finland; InFLAMES Research Flagship Center, Åbo Akademi University and University of Turku, Turku, Finland; Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, the Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Ceres, Building Number 7, De Zaale, 5612 AJ, Eindhoven, the Netherlands.
| |
Collapse
|
5
|
Funk MA, Leitner J, Gerner MC, Hammerler JM, Salzer B, Lehner M, Battin C, Gumpelmair S, Stiasny K, Grabmeier-Pfistershammer K, Steinberger P. Interrogating ligand-receptor interactions using highly sensitive cellular biosensors. Nat Commun 2023; 14:7804. [PMID: 38016944 PMCID: PMC10684770 DOI: 10.1038/s41467-023-43589-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Interactions of membrane-resident proteins are important targets for therapeutic interventions but most methods to study them are either costly, laborious or fail to reflect the physiologic interaction of membrane resident proteins in trans. Here we describe highly sensitive cellular biosensors as a tool to study receptor-ligand pairs. They consist of fluorescent reporter cells that express chimeric receptors harboring ectodomains of cell surface molecules and intracellular signaling domains. We show that a broad range of molecules can be integrated into this platform and we demonstrate its applicability to highly relevant research areas, including the characterization of immune checkpoints and the probing of cells for the presence of receptors or ligands. The platform is suitable to evaluate the interactions of viral proteins with host receptors and to test for neutralization capability of drugs or biological samples. Our results indicate that cellular biosensors have broad utility as a tool to study protein-interactions.
Collapse
Affiliation(s)
- Maximilian A Funk
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria.
| | - Marlene C Gerner
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Jasmin M Hammerler
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Benjamin Salzer
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria
| | - Manfred Lehner
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria
| | - Claire Battin
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Simon Gumpelmair
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Peter Steinberger
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Bertolini M, Wong MS, Mendive-Tapia L, Vendrell M. Smart probes for optical imaging of T cells and screening of anti-cancer immunotherapies. Chem Soc Rev 2023; 52:5352-5372. [PMID: 37376918 PMCID: PMC10424634 DOI: 10.1039/d2cs00928e] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Indexed: 06/29/2023]
Abstract
T cells are an essential part of the immune system with crucial roles in adaptive response and the maintenance of tissue homeostasis. Depending on their microenvironment, T cells can be differentiated into multiple states with distinct functions. This myriad of cellular activities have prompted the development of numerous smart probes, ranging from small molecule fluorophores to nanoconstructs with variable molecular architectures and fluorescence emission mechanisms. In this Tutorial Review, we summarize recent efforts in the design, synthesis and application of smart probes for imaging T cells in tumors and inflammation sites by targeting metabolic and enzymatic biomarkers as well as specific surface receptors. Finally, we briefly review current strategies for how smart probes are employed to monitor the response of T cells to anti-cancer immunotherapies. We hope that this Review may help chemists, biologists and immunologists to design the next generation of molecular imaging probes for T cells and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Marco Bertolini
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Man Sing Wong
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Lorena Mendive-Tapia
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| |
Collapse
|
7
|
Du R, Li L, Ji J, Fan Y. Receptor-Ligand Binding: Effect of Mechanical Factors. Int J Mol Sci 2023; 24:ijms24109062. [PMID: 37240408 DOI: 10.3390/ijms24109062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/20/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Gaining insight into the in situ receptor-ligand binding is pivotal for revealing the molecular mechanisms underlying the physiological and pathological processes and will contribute to drug discovery and biomedical application. An important issue involved is how the receptor-ligand binding responds to mechanical stimuli. This review aims to provide an overview of the current understanding of the effect of several representative mechanical factors, such as tension, shear stress, stretch, compression, and substrate stiffness on receptor-ligand binding, wherein the biomedical implications are focused. In addition, we highlight the importance of synergistic development of experimental and computational methods for fully understanding the in situ receptor-ligand binding, and further studies should focus on the coupling effects of these mechanical factors.
Collapse
Affiliation(s)
- Ruotian Du
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Jing Ji
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
8
|
Pettmann J, Awada L, Różycki B, Huhn A, Faour S, Kutuzov M, Limozin L, Weikl TR, van der Merwe PA, Robert P, Dushek O. Mechanical forces impair antigen discrimination by reducing differences in T-cell receptor/peptide-MHC off-rates. EMBO J 2023; 42:e111841. [PMID: 36484367 PMCID: PMC10068313 DOI: 10.15252/embj.2022111841] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
T cells use their T-cell receptors (TCRs) to discriminate between lower-affinity self and higher-affinity foreign peptide major-histocompatibility-complexes (pMHCs) based on the TCR/pMHC off-rate. It is now appreciated that T cells generate mechanical forces during this process but how force impacts the TCR/pMHC off-rate remains debated. Here, we measured the effect of mechanical force on the off-rate of multiple TCR/pMHC interactions. Unexpectedly, we found that lower-affinity TCR/pMHCs with faster solution off-rates were more resistant to mechanical force (weak slip or catch bonds) than higher-affinity interactions (strong slip bonds). This was confirmed by molecular dynamics simulations. Consistent with these findings, we show that the best-characterized catch bond, involving the OT-I TCR, has a low affinity and an exceptionally fast solution off-rate. Our findings imply that reducing forces on the TCR/pMHC interaction improves antigen discrimination, and we suggest a role for the adhesion receptors CD2 and LFA-1 in force-shielding the TCR/pMHC interaction.
Collapse
Affiliation(s)
| | - Lama Awada
- Laboratoire Adhesion et InflammationAix Marseille University UM 61, INSERM UMRS 1067, CNRS UMR 7333MarseilleFrance
| | | | - Anna Huhn
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Sara Faour
- Laboratoire Adhesion et InflammationAix Marseille University UM 61, INSERM UMRS 1067, CNRS UMR 7333MarseilleFrance
| | - Mikhail Kutuzov
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Laurent Limozin
- Laboratoire Adhesion et InflammationAix Marseille University UM 61, INSERM UMRS 1067, CNRS UMR 7333MarseilleFrance
| | - Thomas R Weikl
- Max Planck Institute of Colloids and InterfacesPotsdamGermany
| | | | - Philippe Robert
- Laboratoire Adhesion et InflammationAix Marseille University UM 61, INSERM UMRS 1067, CNRS UMR 7333MarseilleFrance
- Assistance Publique‐Hôpitaux de MarseilleMarseilleFrance
| | - Omer Dushek
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| |
Collapse
|
9
|
Qin R, An C, Chen W. Physical-Chemical Regulation of Membrane Receptors Dynamics in Viral Invasion and Immune Defense. J Mol Biol 2023; 435:167800. [PMID: 36007627 PMCID: PMC9394170 DOI: 10.1016/j.jmb.2022.167800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 02/04/2023]
Abstract
Mechanical cues dynamically regulate membrane receptors functions to trigger various physiological and pathological processes from viral invasion to immune defense. These cues mainly include various types of dynamic mechanical forces and the spatial confinement of plasma membrane. However, the molecular mechanisms of how they couple with biochemical cues in regulating membrane receptors functions still remain mysterious. Here, we review recent advances in methodologies of single-molecule biomechanical techniques and in novel biomechanical regulatory mechanisms of critical ligand recognition of viral and immune receptors including SARS-CoV-2 spike protein, T cell receptor (TCR) and other co-stimulatory immune receptors. Furthermore, we provide our perspectives of the general principle of how force-dependent kinetics determine the dynamic functions of membrane receptors and of biomechanical-mechanism-driven SARS-CoV-2 neutralizing antibody design and TCR engineering for T-cell-based therapies.
Collapse
Affiliation(s)
- Rui Qin
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Chenyi An
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory for Modern Optical Instrumentation Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
10
|
Han X, Chen Y, Zhang N, Huang C, He G, Li T, Wei M, Song Q, Mo S, Lv Y. Single-cell mechanistic studies of radiation-mediated bystander effects. Front Immunol 2022; 13:849341. [PMID: 36389749 PMCID: PMC9640915 DOI: 10.3389/fimmu.2022.849341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022] Open
Abstract
Ionizing radiation (IR) has been widely used in the diagnosis and treatment of clinical diseases, with radiation therapy (RT) being particularly rapid, but it can induce “bystander effects” that lead to biological responses in non-target cells after their neighboring cells have been irradiated. To help clarify how radiotherapy induces these effects, To help clarify how radiotherapy induces these effects, we analyzed single-cell RNA sequencing data from irradiated intestinal tissues on day 1 (T1 state), day 3 (T3 state), day 7 (T7 state), and day 14 (T14 state) after irradiation, as well as from healthy intestinal tissues (T0 state), to reveal the cellular level, molecular level, and involvement of different time irradiated mouse intestinal tissues in biological signaling pathways. In addition, changes in immune cell subpopulations and myeloid cell subpopulations after different radiation times were further explored, and gene regulatory networks (GRNs) of these cell subpopulations were constructed. Cellular communication between radiation-specific immune cells was explored by cell-to-cell communication events. The results suggest that radiotherapy trigger changes in immune cell subsets, which then reprogram the immune ecosystem and mediate systemic bystander effects. These radiation-specific immune cells participate in a wide range of cell-to-cell communication events. In particular, radiation-specific CD8+T cells appear to be at the core of communication and appear to persist in the body after recovery from radiotherapy, with enrichment analysis showing that radiation-specific CD8+ T cells are associated with ferroptosis. Thus, radiation-specific CD8+ T cells may be involved in cellular ferroptosis-mediated adverse effects caused by RT.
Collapse
Affiliation(s)
- Xueqiong Han
- Department of Oncology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yixuan Chen
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, China
| | - Nan Zhang
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, China
| | - Chengyu Huang
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, China
| | - Guangyao He
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Guangxi Medical University, Nanning, China
| | - Ting Li
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, China
| | - Mengxin Wei
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, China
| | - Qiong Song
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, China
| | - Shaowen Mo
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, China
- *Correspondence: Shaowen Mo, ; Yufeng Lv,
| | - Yufeng Lv
- Department of Oncology, Foresea Life Insurance Guangxi Hospital, Nanning, China
- *Correspondence: Shaowen Mo, ; Yufeng Lv,
| |
Collapse
|
11
|
Arricca M, Salvadori A, Bonanno C, Serpelloni M. Modeling Receptor Motility along Advecting Lipid Membranes. MEMBRANES 2022; 12:membranes12070652. [PMID: 35877855 PMCID: PMC9317916 DOI: 10.3390/membranes12070652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 11/24/2022]
Abstract
This work aims to overview multiphysics mechanobiological computational models for receptor dynamics along advecting cell membranes. Continuum and statistical models of receptor motility are the two main modeling methodologies identified in reviewing the state of the art. Within the former modeling class, a further subdivision based on different biological purposes and processes of proteins’ motion is recognized; cell adhesion, cell contractility, endocytosis, and receptor relocations on advecting membranes are the most relevant biological processes identified in which receptor motility is pivotal. Numerical and/or experimental methods and approaches are highlighted in the exposure of the reviewed works provided by the literature, pertinent to the topic of the present manuscript. With a main focus on the continuum models of receptor motility, we discuss appropriate multiphyisics laws to model the mass flux of receptor proteins in the reproduction of receptor relocation and recruitment along cell membranes to describe receptor–ligand chemical interactions, and the cell’s structural response. The mass flux of receptor modeling is further supported by a discussion on the methodology utilized to evaluate the protein diffusion coefficient developed over the years.
Collapse
Affiliation(s)
- Matteo Arricca
- The Mechanobiology Research Center, University of Brescia (UNIBS), 25123 Brescia, Italy; (M.A.); (C.B.); (M.S.)
- Department of Mechanical and Industrial Engineering, Università degli Studi di Brescia, via Branze 38, 25123 Brescia, Italy
| | - Alberto Salvadori
- The Mechanobiology Research Center, University of Brescia (UNIBS), 25123 Brescia, Italy; (M.A.); (C.B.); (M.S.)
- Department of Mechanical and Industrial Engineering, Università degli Studi di Brescia, via Branze 38, 25123 Brescia, Italy
- Correspondence:
| | - Claudia Bonanno
- The Mechanobiology Research Center, University of Brescia (UNIBS), 25123 Brescia, Italy; (M.A.); (C.B.); (M.S.)
- Department of Civil, Environmental, Architectural Engineering and Mathematics, Università degli Studi di Brescia, via Branze 43, 25123 Brescia, Italy
| | - Mattia Serpelloni
- The Mechanobiology Research Center, University of Brescia (UNIBS), 25123 Brescia, Italy; (M.A.); (C.B.); (M.S.)
- Department of Mechanical and Industrial Engineering, Università degli Studi di Brescia, via Branze 38, 25123 Brescia, Italy
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Rahmaninejad H, Pace T, Chun BJ, Kekenes-Huskey PM. Crowding within synaptic junctions influences the degradation of nucleotides by CD39 and CD73 ectonucleotidases. Biophys J 2022; 121:309-318. [PMID: 34922916 PMCID: PMC8790186 DOI: 10.1016/j.bpj.2021.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/01/2021] [Accepted: 12/07/2021] [Indexed: 01/21/2023] Open
Abstract
Synapsed cells can communicate using exocytosed nucleotides like adenosine triphosphate (ATP). Ectonucleotidases localized to synaptic junctions degrade nucleotides into metabolites like adenosine monophosphate (AMP) or adenosine. Oftentimes nucleotide degradation occurs in a sequential manner, of which ATP degradation by CD39 and CD73 is a representative example. Here, CD39 first converts ATP and adenosine diphosphate (ADP) into AMP, after which AMP is dephosphorylated into adenosine by CD73. Hence, the concerted activity of CD39 and CD73 can help shape cellular responses to extracellular ATP. In a previous study, we demonstrated that coupled CD39 and CD73 activity within synapse-like junctions is strongly controlled by the enzymes' co-localization, their surface charge densities, and the electrostatic potential of the surrounding cell membranes. In this study, we demonstrate that crowders within synaptic junctions, which can include globular proteins like cytokines and membrane-bound proteins, impact coupled CD39 and CD73 ectonucleotidase activity and, in turn, the availability of intrasynapse ATP. Specifically, we developed a spatially explicit, reaction-diffusion model for the coupled conversion of ATP → AMP and AMP → adenosine in a model synaptic junction with crowders that is solved via the finite element method. Our modeling results suggest that the association rate for ATP to CD39 is strongly influenced by the density of intrasynaptic protein crowders, as increasing crowder density generally suppressed ATP association kinetics. Much of this suppression can be rationalized based on a loss of configurational entropy. The surface charges of crowders can further influence the association rate, with the surprising result that favorable crowder-nucleotide electrostatic interactions can yield CD39 association rates that are faster than crowder-free configurations. However, attractive crowder-nucleotide interactions decrease the rate and efficiency of adenosine production, which in turn increases the availability of ATP and AMP within the synapse relative to crowder-free configurations. These findings highlight how CD39 and CD73 ectonucleotidase activity, electrostatics, and crowding within synapses influence the availability of nucleotides for intercellular communication.
Collapse
Affiliation(s)
- Hadi Rahmaninejad
- Department of Physics, Virginia Tech, Blacksburg,Corresponding author
| | - Tom Pace
- Department of Cell & Molecular Physiology, Loyola University Chicago, Chicago,Corresponding author
| | - Byeong Jae Chun
- Department of Cell & Molecular Physiology, Loyola University Chicago, Chicago
| | | |
Collapse
|
15
|
Jian C, Wei L, Mo R, Li R, Liang L, Chen L, Zou C, Meng Y, Liu Y, Zou D. Microglia Mediate the Occurrence and Development of Alzheimer's Disease Through Ligand-Receptor Axis Communication. Front Aging Neurosci 2021; 13:731180. [PMID: 34616287 PMCID: PMC8488208 DOI: 10.3389/fnagi.2021.731180] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/01/2021] [Indexed: 01/23/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease. Its onset is insidious and its progression is slow, making diagnosis difficult. In addition, its underlying molecular and cellular mechanisms remain unclear. In this study, clustering analysis was performed on single-cell RNA sequencing (scRNA-seq) data from the prefrontal cortex of 48 AD patients. Each sample module was identified to be a specific AD cell type, eight main brain cell types were identified, and the dysfunctional evolution of each cell type was further explored by pseudo-time analysis. Correlation analysis was then used to explore the relationship between AD cell types and pathological characteristics. In particular, intercellular communication between neurons and glial cells in AD patients was investigated by cell communication analysis. In patients, neuronal cells and glial cells significantly correlated with pathological features, and glial cells appear to play a key role in the development of AD through ligand-receptor axis communication. Marker genes involved in communication between these two cell types were identified using five types of modeling: logistic regression, multivariate logistic regression, least absolute shrinkage and selection operator (LASSO) and support vector machine (SVM). LASSO modeling identified CXCR4, EGFR, MAP4K4, and IGF1R as key genes in this communication. Our results support the idea that microglia play a role in the occurrence and development of AD through ligand-receptor axis communication. In particular, our analyses identify CXCR4, EGFR, MAP4K4, and IGF1R as potential biomarkers and therapeutic targets in AD.
Collapse
Affiliation(s)
- Chongdong Jian
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Lei Wei
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ruikang Mo
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rongjie Li
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lucong Liang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liechun Chen
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chun Zou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Youshi Meng
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ying Liu
- Department of General Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Geriatrics, The First People’s Hospital of Nanning, Nanning, China
| | - Donghua Zou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
16
|
Nappi F, Iervolino A, Avtaar Singh SS. COVID-19 Pathogenesis: From Molecular Pathway to Vaccine Administration. Biomedicines 2021; 9:903. [PMID: 34440107 PMCID: PMC8389702 DOI: 10.3390/biomedicines9080903] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022] Open
Abstract
The Coronavirus 2 (SARS-CoV-2) infection is a global pandemic that has affected millions of people worldwide. The advent of vaccines has permitted some restitution. Aside from the respiratory complications of the infection, there is also a thrombotic risk attributed to both the disease and the vaccine. There are no reliable data for the risk of thromboembolism in SARS-CoV-2 infection in patients managed out of the hospital setting. A literature review was performed to identify the pathophysiological mechanism of thrombosis from the SARS-CoV-2 infection including the role of Angiotensin-Converting Enzyme receptors. The impact of the vaccine and likely mechanisms of thrombosis following vaccination were also clarified. Finally, the utility of the vaccines available against the multiple variants is also highlighted. The systemic response to SARS-CoV-2 infection is still relatively poorly understood, but several risk factors have been identified. The roll-out of the vaccines worldwide has also allowed the lifting of lockdown measures and a reduction in the spread of the disease. The experience of the SARS-CoV-2 infection, however, has highlighted the crucial role of epidemiological research and the need for ongoing studies within this field.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| | - Adelaide Iervolino
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, 00168 Rome, Italy;
| | | |
Collapse
|
17
|
Zhovmer AS, Chandler M, Manning A, Afonin KA, Tabdanov ED. Programmable DNA-augmented hydrogels for controlled activation of human lymphocytes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102442. [PMID: 34284132 DOI: 10.1016/j.nano.2021.102442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/25/2021] [Accepted: 07/07/2021] [Indexed: 12/25/2022]
Abstract
Contractile forces within the planar interface between T cell and antigen-presenting surface mechanically stimulate T cell receptors (TCR) in the mature immune synapses. However, the origin of mechanical stimulation during the initial, i.e., presynaptic, microvilli-based TCR activation in the course of immune surveillance remains unknown and new tools to help address this problem are needed. In this work, we develop nucleic acid nanoassembly (NAN)-based technology for functionalization of hydrogels using isothermal toehold-mediated reassociation of RNA/DNA heteroduplexes. Resulting platform allows for regulation with NAN linkers of 3D force momentum along the TCR mechanical axis, whereas hydrogels contribute to modulation of 2D shear modulus. By utilizing different lengths of NAN linkers conjugated to polyacrylamide gels of different shear moduli, we demonstrate an efficient capture of human T lymphocytes and tunable activation of TCR, as confirmed by T-cell spreading and pY foci.
Collapse
Affiliation(s)
- Alexander S Zhovmer
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| | - Morgan Chandler
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Alexis Manning
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Kirill A Afonin
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA.
| | - Erdem D Tabdanov
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
18
|
Göhring J, Kellner F, Schrangl L, Platzer R, Klotzsch E, Stockinger H, Huppa JB, Schütz GJ. Temporal analysis of T-cell receptor-imposed forces via quantitative single molecule FRET measurements. Nat Commun 2021; 12:2502. [PMID: 33947864 PMCID: PMC8096839 DOI: 10.1038/s41467-021-22775-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 03/25/2021] [Indexed: 02/01/2023] Open
Abstract
Mechanical forces acting on ligand-engaged T-cell receptors (TCRs) have previously been implicated in T-cell antigen recognition, yet their magnitude, spread, and temporal behavior are still poorly defined. We here report a FRET-based sensor equipped either with a TCR-reactive single chain antibody fragment or peptide-loaded MHC, the physiological TCR-ligand. The sensor was tethered to planar glass-supported lipid bilayers (SLBs) and informed most directly on the magnitude and kinetics of TCR-imposed forces at the single molecule level. When confronting T-cells with gel-phase SLBs we observed both prior and upon T-cell activation a single, well-resolvable force-peak of approximately 5 pN and force loading rates on the TCR of 1.5 pN per second. When facing fluid-phase SLBs instead, T-cells still exerted tensile forces yet of threefold reduced magnitude and only prior to but not upon activation.
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
| | - Florian Kellner
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - René Platzer
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Enrico Klotzsch
- Institute of Applied Physics, TU Wien, Vienna, Austria
- Laboratory of Applied Mechanobiology, Department for Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Institute of Biology, Experimental Biophysics/ Mechanobiology, Humboldt Universität zu Berlin, Berlin, Germany
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes B Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | | |
Collapse
|
19
|
Porębska N, Poźniak M, Matynia A, Żukowska D, Zakrzewska M, Otlewski J, Opaliński Ł. Galectins as modulators of receptor tyrosine kinases signaling in health and disease. Cytokine Growth Factor Rev 2021; 60:89-106. [PMID: 33863623 DOI: 10.1016/j.cytogfr.2021.03.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022]
Abstract
Receptor tyrosine kinases (RTKs) constitute a large group of cell surface proteins that mediate communication of cells with extracellular environment. RTKs recognize external signals and transfer information to the cell interior, modulating key cellular activities, like metabolism, proliferation, motility, or death. To ensure balanced stream of signals the activity of RTKs is tightly regulated by numerous mechanisms, including receptor expression and degradation, ligand specificity and availability, engagement of co-receptors, cellular trafficking of the receptors or their post-translational modifications. One of the most widespread post-translational modifications of RTKs is glycosylation of their extracellular domains. The sugar chains attached to RTKs form a new layer of information, so called glyco-code that is read by galectins, carbohydrate binding proteins. Galectins are family of fifteen lectins implicated in immune response, inflammation, cell division, motility and death. The versatility of cellular activities attributed to galectins is a result of their high abundance and diversity of their cellular targets. A various sugar specificity of galectins and the differential ability of galectin family members to form oligomers affect the spatial distribution and the function of their cellular targets. Importantly, galectins and RTKs are tightly linked to the development, progression and metastasis of various cancers. A growing number of studies points on the close cooperation between RTKs and galectins in eliciting specific cellular responses. This review focuses on the identified complexes between galectins and RTK members and discusses their relevance for the cell physiology both in healthy tissues and in cancer.
Collapse
Affiliation(s)
- Natalia Porębska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Marta Poźniak
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Aleksandra Matynia
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Dominika Żukowska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland.
| |
Collapse
|
20
|
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
|
21
|
Razvag Y, Neve-Oz Y, Sajman J, Yakovian O, Reches M, Sherman E. T Cell Activation through Isolated Tight Contacts. Cell Rep 2020; 29:3506-3521.e6. [PMID: 31825832 DOI: 10.1016/j.celrep.2019.11.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 08/12/2019] [Accepted: 11/06/2019] [Indexed: 01/01/2023] Open
Abstract
T cells engage antigen-presenting cells in search for cognate antigens via dynamic cell protrusions before forming a tight immune synapse. The spatiotemporal events that may lead to rapid TCR triggering and signal amplification in microvilli-driven isolated contacts, and in subsequent, more uniform contacts, remain poorly understood. Here, we combined interference-reflectance microscopy and single-molecule localization microscopy in live cells to resolve TCR-dependent signaling at tight cell contacts. We show that early contacts are sufficient for robust TCR triggering and ZAP-70 recruitment. With cell spreading, TCR activation and ZAP-70 recruitment increase and shift to the edges of the growing tight contacts. CD45 segregates from TCR at tight contacts and is enriched at high local curvature membrane. Surprisingly, cortical actin and LFA localized at contact regions of intermediate tightness. Our results show in molecular detail the roles of early and tight T cell contacts in T cell activation, as both sensing and decision-making entities.
Collapse
Affiliation(s)
- Yair Razvag
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel; Institute of Chemistry and The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yair Neve-Oz
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel
| | - Julia Sajman
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel
| | - Oren Yakovian
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel
| | - Meital Reches
- Institute of Chemistry and The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel.
| |
Collapse
|
22
|
Carotenuto AR, Lunghi L, Piccolo V, Babaei M, Dayal K, Pugno N, Zingales M, Deseri L, Fraldi M. Mechanobiology predicts raft formations triggered by ligand-receptor activity across the cell membrane. JOURNAL OF THE MECHANICS AND PHYSICS OF SOLIDS 2020; 141:103974. [PMID: 32461703 PMCID: PMC7243794 DOI: 10.1016/j.jmps.2020.103974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 05/05/2023]
Abstract
Clustering of ligand-binding receptors of different types on thickened isles of the cell membrane, namely lipid rafts, is an experimentally observed phenomenon. Although its influence on cell's response is deeply investigated, the role of the coupling between mechanical processes and multiphysics involving the active receptors and the surrounding lipid membrane during ligand-binding has not yet been understood. Specifically, the focus of this work is on G-protein-coupled receptors (GPCRs), the widest group of transmembrane proteins in animals, which regulate specific cell processes through chemical signalling pathways involving a synergistic balance between the cyclic Adenosine Monophosphate (cAMP) produced by active GPCRs in the intracellular environment and its efflux, mediated by the Multidrug Resistance Proteins (MRPs) transporters. This paper develops a multiphysics approach based on the interplay among energetics, multiscale geometrical changes and mass balance of species, i.e. active GPCRs and MRPs, including diffusion and kinetics of binding and unbinding. Because the obtained energy depends upon both the kinematics and the changes of species densities, balance of mass and of linear momentum are coupled and govern the space-time evolution of the cell membrane. The mechanobiology involving remodelling and change of lipid ordering of the cell membrane allows to predict dynamics of transporters and active receptors -in full agreement with experimentally observed cAMP levels- and how the latter trigger rafts formation and cluster on such sites. Within the current scientific debate on Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) and on the basis of the ascertained fact that lipid rafts often serve as an entry port for viruses, it is felt that approaches accounting for strong coupling among mechanobiological aspects could even turn helpful in better understanding membrane-mediated phenomena such as COVID-19 virus-cell interaction.
Collapse
Affiliation(s)
- Angelo R. Carotenuto
- Department of Structures for Engineering and Architecture, University of Napoli “Federico II”, Italy
| | - Laura Lunghi
- Smiling International School, formerly at the Department of Life Sciences and Biotech., University of Ferrara, Italy
| | - Valentina Piccolo
- Department of Civil, Environmental and Mechanical Engineering, University of Trento, Italy
| | - Mahnoush Babaei
- Department of Civil and Environmental Engineering, Department of Mechanical Engineering, Carnegie Mellon, USA
| | - Kaushik Dayal
- Department of Civil and Environmental Engineering, Department of Mechanical Engineering, Carnegie Mellon, USA
| | - Nicola Pugno
- Department of Civil, Environmental and Mechanical Engineering, University of Trento, Italy
- Laboratory of Bio-inspired, Bionic, Nano, Meta Materials & Mechanics, Department of Civil, Environmental and Mechanical Engineering, University of Trento, Via Mesiano, 77, Trento 38123, Italy
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Massimiliano Zingales
- Dipartimento di Ingegneria, Universitàdi Palermo, viale delle Scienze ed.8, 90128 Palermo, Italy
| | - Luca Deseri
- Department of Civil, Environmental and Mechanical Engineering, University of Trento, Italy
- Department of Civil and Environmental Engineering, Department of Mechanical Engineering, Carnegie Mellon, USA
- Department of Mechanical Engineering and Material Sciences, SSoE, University of Pittsburgh USA
- Department of Nanomedicine, The Houston Methodist Research Institute, USA
| | - Massimiliano Fraldi
- Department of Structures for Engineering and Architecture, University of Napoli “Federico II”, Italy
| |
Collapse
|
23
|
Abstract
T cells initiate and regulate adaptive immune responses that can clear infections. To do this, they use their T cell receptors (TCRs) to continually scan the surfaces of other cells for cognate peptide antigens presented on major histocompatibility complexes (pMHCs). Experimental work has established that as few 1-10 pMHCs are sufficient to activate T cells. This sensitivity is remarkable in light of a number of factors, including the observation that the TCR and pMHC are short molecules relative to highly abundant long surface molecules, such as CD45, that can hinder initial binding, and moreover, the TCR/pMHC interaction is of weak affinity with solution lifetimes of approximately 1 second. Here, we review experimental and mathematical work that has contributed to uncovering molecular mechanisms of T cell sensitivity. We organize the mechanisms by where they act in the pathway to activate T cells, namely mechanisms that (a) promote TCR/pMHC binding, (b) induce rapid TCR signaling, and (c) amplify TCR signaling. We discuss work showing that high sensitivity reduces antigen specificity unless molecular feedbacks are invoked. We conclude by summarizing a number of open questions.
Collapse
Affiliation(s)
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Liu K, Chu B, Newby J, Read EL, Lowengrub J, Allard J. Hydrodynamics of transient cell-cell contact: The role of membrane permeability and active protrusion length. PLoS Comput Biol 2019; 15:e1006352. [PMID: 31022168 PMCID: PMC6504115 DOI: 10.1371/journal.pcbi.1006352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 05/07/2019] [Accepted: 02/01/2019] [Indexed: 11/19/2022] Open
Abstract
In many biological settings, two or more cells come into physical contact to form a cell-cell interface. In some cases, the cell-cell contact must be transient, forming on timescales of seconds. One example is offered by the T cell, an immune cell which must attach to the surface of other cells in order to decipher information about disease. The aspect ratio of these interfaces (tens of nanometers thick and tens of micrometers in diameter) puts them into the thin-layer limit, or "lubrication limit", of fluid dynamics. A key question is how the receptors and ligands on opposing cells come into contact. What are the relative roles of thermal undulations of the plasma membrane and deterministic forces from active filopodia? We use a computational fluid dynamics algorithm capable of simulating 10-nanometer-scale fluid-structure interactions with thermal fluctuations up to seconds- and microns-scales. We use this to simulate two opposing membranes, variously including thermal fluctuations, active forces, and membrane permeability. In some regimes dominated by thermal fluctuations, proximity is a rare event, which we capture by computing mean first-passage times using a Weighted Ensemble rare-event computational method. Our results demonstrate a parameter regime in which the time it takes for an active force to drive local contact actually increases if the cells are being held closer together (e.g., by nonspecific adhesion), a phenomenon we attribute to the thin-layer effect. This leads to an optimal initial cell-cell separation for fastest receptor-ligand binding, which could have relevance for the role of cellular protrusions like microvilli. We reproduce a previous experimental observation that fluctuation spatial scales are largely unaffected, but timescales are dramatically slowed, by the thin-layer effect. We also find that membrane permeability would need to be above physiological levels to abrogate the thin-layer effect.
Collapse
Affiliation(s)
- Kai Liu
- Department of Mathematics, University of California Irvine, Irvine, California, United States of America
- Center for Mathematical Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Brian Chu
- Department of Chemical Engineering and Materials Science, University of California Irvine, Irvine, California, United States of America
| | - Jay Newby
- Department of Mathematics, University of Alberta, Edmonton, Alberta, Canada
| | - Elizabeth L. Read
- Department of Chemical Engineering and Materials Science, University of California Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California Irvine, Irvine, California, United States of America
| | - John Lowengrub
- Department of Mathematics, University of California Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California Irvine, Irvine, California, United States of America
| | - Jun Allard
- Department of Mathematics, University of California Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California Irvine, Irvine, California, United States of America
- Department of Physics and Astronomy, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
25
|
Jing H, Sinha S, Sachar HS, Das S. Interactions of gold and silica nanoparticles with plasma membranes get distinguished by the van der Waals forces: Implications for drug delivery, imaging, and theranostics. Colloids Surf B Biointerfaces 2019; 177:433-439. [PMID: 30798064 DOI: 10.1016/j.colsurfb.2019.01.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/25/2019] [Accepted: 01/30/2019] [Indexed: 12/31/2022]
Abstract
Making a nanoparticle (NP) approach and interact with a plasma membrane (PM) through the receptor-ligand interaction is key for applications like targeted drug delivery, cellular imaging, and theranostics. In this paper, we show that the van der Waals (vdW) interactions dominate the electrostatics ensuring that a gold NP approached the PM more spontaneously as compared to a silica NP. The negative σ (charge density) of a PM induces a negative electrostatic potential at the surface of the approaching gold NP and the silica NP; however, there is very little difference between these induced values due to a small electric double layer at the physiological salt concentration (c∞). Hence there is very little difference in the electrostatic repulsion between the two cases, while the PM-NP vdW attraction is much more for the gold NP as a result of a larger Hamaker constant. Therefore, in comparison to the gold NP, the silica NP would (a) undergo a promotion of the specific adhesion and a prevention of the non-specific adhesion simultaneously for a larger σ - c∞ phase space including the physiological conditions, (b) necessitate a larger length of the ligands to trigger spontaneous receptor-ligand interactions, and (c) require a larger driving force for force-driven receptor-ligand interactions.
Collapse
Affiliation(s)
- Haoyuan Jing
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA
| | - Shayandev Sinha
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA
| | - Harnoor Singh Sachar
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA
| | - Siddhartha Das
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
26
|
Neve-Oz Y, Sajman J, Razvag Y, Sherman E. InterCells: A Generic Monte-Carlo Simulation of Intercellular Interfaces Captures Nanoscale Patterning at the Immune Synapse. Front Immunol 2018; 9:2051. [PMID: 30254635 PMCID: PMC6141710 DOI: 10.3389/fimmu.2018.02051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/20/2018] [Indexed: 12/03/2022] Open
Abstract
Molecular interactions across intercellular interfaces serve to convey information between cells and to trigger appropriate cell functions. Examples include cell development and growth in tissues, neuronal and immune synapses (ISs). Here, we introduce an agent-based Monte-Carlo simulation of user-defined cellular interfaces. The simulation allows for membrane molecules, embedded at intercellular contacts, to diffuse and interact, while capturing the topography and energetics of the plasma membranes of the interface. We provide a detailed example related to pattern formation in the early IS. Using simulation predictions and three-color single molecule localization microscopy (SMLM), we detected the intricate mutual patterning of T cell antigen receptors (TCRs), integrins and glycoproteins in early T cell contacts with stimulating coverslips. The simulation further captures the dynamics of the patterning under the experimental conditions and at the IS with antigen presenting cells (APCs). Thus, we provide a generic tool for simulating realistic cell-cell interfaces, which can be used for critical hypothesis testing and experimental design in an iterative manner.
Collapse
Affiliation(s)
- Yair Neve-Oz
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| | - Julia Sajman
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| | - Yair Razvag
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
27
|
Gp41 dynamically interacts with the TCR in the immune synapse and promotes early T cell activation. Sci Rep 2018; 8:9747. [PMID: 29950577 PMCID: PMC6021400 DOI: 10.1038/s41598-018-28114-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/14/2018] [Indexed: 12/21/2022] Open
Abstract
The HIV-1 glycoprotein gp41 critically mediates CD4+ T-cell infection by HIV-1 during viral entry, assembly, and release. Although multiple immune-regulatory activities of gp41 have been reported, the underlying mechanisms of these activities remain poorly understood. Here we employed multi-colour single molecule localization microscopy (SMLM) to resolve interactions of gp41 proteins with cellular proteins at the plasma membrane (PM) of fixed and live CD4+ T-cells with resolution of ~20–30 nm. We observed that gp41 clusters dynamically associated with the T cell antigen receptor (TCR) at the immune synapse upon TCR stimulation. This interaction, confirmed by FRET, depended on the virus clone, was reduced by the gp41 ectodomain in tight contacts, and was completely abrogated by mutation of the gp41 transmembrane domain. Strikingly, gp41 preferentially colocalized with phosphorylated TCRs at the PM of activated T-cells and promoted TCR phosphorylation. Gp41 expression also resulted in enhanced CD69 upregulation, and in massive cell death after 24–48 hrs. Our results shed new light on HIV-1 assembly mechanisms at the PM of host T-cells and its impact on TCR stimulation.
Collapse
|
28
|
Al-Aghbar MA, Chu YS, Chen BM, Roffler SR. High-Affinity Ligands Can Trigger T Cell Receptor Signaling Without CD45 Segregation. Front Immunol 2018; 9:713. [PMID: 29686683 PMCID: PMC5900011 DOI: 10.3389/fimmu.2018.00713] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/22/2018] [Indexed: 11/13/2022] Open
Abstract
How T cell receptors (TCRs) are triggered to start signaling is still not fully understood. It has been proposed that segregation of the large membrane tyrosine phosphatase CD45 from engaged TCRs initiates signaling by favoring phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs) in the cytoplasmic domains of CD3 molecules. However, whether CD45 segregation is important to initiate triggering is still uncertain. We examined CD45 segregation from TCRs engaged to anti-CD3 scFv with high or low affinity and with defined molecular lengths on glass-supported lipid bilayers using total internal reflection microscopy. Both short and elongated high-affinity anti-CD3 scFv effectively induced similar calcium mobilization, Zap70 phosphorylation, and cytokine secretion in Jurkat T cells but CD45 segregated from activated TCR microclusters significantly less for elongated versus short anti-CD3 ligands. In addition, at early times, triggering cells with both high and low affinity elongated anti-CD3 scFv resulted in similar degrees of CD3 co-localization with CD45, but only the high-affinity scFv induced T cell activation. The lack of correlation between CD45 segregation and early markers of T cell activation suggests that segregation of CD45 from engaged TCRs is not mandatory for initial triggering of TCR signaling by elongated high-affinity ligands.
Collapse
Affiliation(s)
- Mohammad Ameen Al-Aghbar
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University, Academia Sinica, Taipei, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Yeh-Shiu Chu
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
29
|
Nanoscale kinetic segregation of TCR and CD45 in engaged microvilli facilitates early T cell activation. Nat Commun 2018; 9:732. [PMID: 29467364 PMCID: PMC5821895 DOI: 10.1038/s41467-018-03127-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 01/18/2018] [Indexed: 11/08/2022] Open
Abstract
T cells have a central function in mounting immune responses. However, mechanisms of their early activation by cognate antigens remain incompletely understood. Here we use live-cell multi-colour single-molecule localization microscopy to study the dynamic separation between TCRs and CD45 glycoprotein phosphatases in early cell contacts under TCR-activating and non-activating conditions. Using atomic force microscopy, we identify these cell contacts with engaged microvilli and characterize their morphology, rigidity and dynamics. Physical modelling and simulations of the imaged cell interfaces quantitatively capture the TCR-CD45 separation. Surprisingly, TCR phosphorylation negatively correlates with TCR-CD45 separation. These data support a refined kinetic-segregation model. First, kinetic-segregation occurs within seconds from TCR activation in engaged microvilli. Second, TCRs should be segregated, yet not removed too far, from CD45 for their optimal and localized activation within clusters. Our combined imaging and computational approach prove an important tool in the study of dynamic protein organization in cell interfaces.
Collapse
|
30
|
Catch Bonds at T Cell Interfaces: Impact of Surface Reorganization and Membrane Fluctuations. Biophys J 2017; 113:120-131. [PMID: 28700910 DOI: 10.1016/j.bpj.2017.05.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/07/2017] [Accepted: 05/19/2017] [Indexed: 01/30/2023] Open
Abstract
Catch bonds are characterized by average lifetimes that initially increase with increasing tensile force. Recently, they have been implicated in T cell activation, where small numbers of antigenic receptor-ligand bonds at a cell-cell interface can stimulate a T cell. Here, we use computational methods to investigate small numbers of bonds at the interface between two membranes. We characterize the time-dependent forces on the bonds in response to changes in the membrane shape and the organization of other surface molecules. We then determine the distributions of bond lifetimes using recent force-dependent lifetime data for T cell receptors bound to various ligands. Strong agonists, which exhibit catch bond behavior, are markedly more likely to remain intact than an antagonist whose average lifetime decreases with increasing force. Thermal fluctuations of the membrane shape enhance the decay of the average force on a bond, but also lead to fluctuations of the force. These fluctuations promote bond rupture, but the effect is buffered by catch bonds. When more than one bond is present, the bonds experience reduced average forces that depend on their relative positions, leading to changes in bond lifetimes. Our results highlight the importance of force-dependent binding kinetics when bonds experience time-dependent and fluctuating forces, as well as potential consequences of collective bond behavior relevant to T cell activation.
Collapse
|
31
|
Mechanosensing in the immune response. Semin Cell Dev Biol 2017; 71:137-145. [PMID: 28830744 DOI: 10.1016/j.semcdb.2017.08.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 01/16/2023]
Abstract
Cells have a remarkable ability to sense and respond to the mechanical properties of their environment. Mechanosensing is essential for many phenomena, ranging from cell movements and tissue rearrangements to cell differentiation and the immune response. Cells of the immune system get activated when membrane receptors bind to cognate antigen on the surface of antigen presenting cells. Both T and B lymphocyte signaling has been shown to be responsive to physical forces and mechanical cues. Cytoskeletal forces exerted by cells likely mediate this mechanical modulation. Here, we discuss recent advances in the field of immune cell mechanobiology at the molecular and cellular scale.
Collapse
|
32
|
Cell Surface Mechanochemistry and the Determinants of Bleb Formation, Healing, and Travel Velocity. Biophys J 2016; 110:1636-1647. [PMID: 27074688 DOI: 10.1016/j.bpj.2016.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/07/2016] [Accepted: 03/07/2016] [Indexed: 11/21/2022] Open
Abstract
Blebs are pressure-driven cell protrusions implicated in cellular functions such as cell division, apoptosis, and cell motility, including motility of protease-inhibited cancer cells. Because of their mechanical nature, blebs inform us about general cell-surface mechanics, including membrane dynamics, pressure propagation throughout the cytoplasm, and the architecture and dynamics of the actin cortex. Mathematical models including detailed fluid dynamics have previously been used to understand bleb expansion. Here, we develop mathematical models in two and three dimensions on longer timescales that recapitulate the full bleb life cycle, including both expansion and healing by cortex reformation, in terms of experimentally accessible biophysical parameters such as myosin contractility, osmotic pressure, and turnover of actin and ezrin. The model provides conditions under which blebbing occurs, and naturally gives rise to traveling blebs. The model predicts conditions under which blebs travel or remain stationary, as well as the bleb traveling velocity, a quantity that has remained elusive in previous models. As previous studies have used blebs as reporters of membrane tension and pressure dynamics within the cell, we have used our system to investigate various pressure equilibration models and dynamic, nonuniform membrane tension to account for the shape of a traveling bleb. We also find that traveling blebs tend to expand in all directions unless otherwise constrained. One possible constraint could be provided by spatial heterogeneity in, for example, adhesion density.
Collapse
|
33
|
Newby J, Allard J. First-Passage Time to Clear the Way for Receptor-Ligand Binding in a Crowded Environment. PHYSICAL REVIEW LETTERS 2016; 116:128101. [PMID: 27058103 DOI: 10.1103/physrevlett.116.128101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Indexed: 06/05/2023]
Abstract
Certain biological reactions, such as receptor-ligand binding at cell-cell interfaces and macromolecules binding to biopolymers, require many smaller molecules crowding a reaction site to be cleared. Examples include the T-cell interface, a key player in immunological information processing. Diffusion sets a limit for such cavitation to occur spontaneously, thereby defining a time scale below which active mechanisms must take over. We consider N independent diffusing particles in a closed domain, containing a subregion with N_{0} particles, on average. We investigate the time until the subregion is empty, allowing a subsequent reaction to proceed. The first-passage time is computed using an efficient exact simulation algorithm and an asymptotic approximation in the limit that cavitation is rare. In this limit, we find that the mean first-passage time is subexponential, T∝e^{N_{0}}/N_{0}^{2}. For the case of T-cell receptors, we find that stochastic cavitation is exceedingly slow, 10^{9} s at physiological densities; however, it can be accelerated to occur within 5 s with only a fourfold dilution.
Collapse
Affiliation(s)
- Jay Newby
- Department of Mathematics, University of North Carolina, Chapel Hill, 329 Phillips Hall, Chapel Hill, North Carolina 27599, USA
| | - Jun Allard
- Department of Mathematics, University of California, Irvine, 340 Rowland Hall, Irvine, California 92697, USA
| |
Collapse
|
34
|
Carlson A, Mahadevan L. Elastohydrodynamics and Kinetics of Protein Patterning in the Immunological Synapse. PLoS Comput Biol 2015; 11:e1004481. [PMID: 26699430 PMCID: PMC4689476 DOI: 10.1371/journal.pcbi.1004481] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 07/30/2015] [Indexed: 11/19/2022] Open
Abstract
We propose a minimal mathematical model for the physical basis of membrane protein patterning in the immunological synapse (IS), which encompass membrane mechanics, protein binding kinetics and motion, and fluid flow in the synaptic cleft. Our theory leads to simple predictions for the spatial and temporal scales of protein cluster formation, growth and arrest as a function of membrane stiffness, rigidity and kinetics of the adhesive proteins, and the fluid flow in the synaptic cleft. Numerical simulations complement these scaling laws by quantifying the nucleation, growth and stabilization of proteins domains on the size of the cell. Direct comparison with experiment shows that passive elastohydrodynamics and kinetics of protein binding in the synaptic cleft can describe the short-time formation and organization of protein clusters, without evoking any active processes in the cytoskeleton. Despite the apparent complexity of the process, our analysis shows that just two dimensionless parameters characterize the spatial and temporal evolution of the protein pattern: a ratio of membrane elasticity to protein stiffness, and the ratio of a hydrodynamic time scale for fluid flow relative to the protein binding rate. A simple phase diagram encompasses the variety of patterns that can arise.
Collapse
Affiliation(s)
- Andreas Carlson
- School of Engineering and Applied Sciences, Kavli Institute for Bionano Science and Technology, and Wyss Institute, Harvard University, Cambridge, United States of America
| | - L. Mahadevan
- School of Engineering and Applied Sciences, Kavli Institute for Bionano Science and Technology, and Wyss Institute, Harvard University, Cambridge, United States of America
- Departments of Physics, and Organismic and Evolutionary Biology, Harvard University, Cambridge, United States of America
- * E-mail:
| |
Collapse
|
35
|
Liu B, Chen W, Natarajan K, Li Z, Margulies DH, Zhu C. The cellular environment regulates in situ kinetics of T-cell receptor interaction with peptide major histocompatibility complex. Eur J Immunol 2015; 45:2099-110. [PMID: 25944482 DOI: 10.1002/eji.201445358] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/17/2015] [Accepted: 04/30/2015] [Indexed: 11/12/2022]
Abstract
T cells recognize antigens at the two-dimensional (2D) interface with antigen-presenting cells (APCs), which trigger T-cell effector functions. T-cell functional outcomes correlate with 2D kinetics of membrane-embedded T-cell receptors (TCRs) binding to surface-tethered peptide-major histocompatibility complex molecules (pMHCs). However, most studies have measured TCR-pMHC kinetics for recombinant TCRs in 3D by surface plasmon resonance, which differs drastically from 2D measurements. Here, we compared pMHC dissociation from native TCR on the T-cell surface to recombinant TCR immobilized on glass surface or in solution. Force on TCR-pMHC bonds regulated their lifetimes differently for native than recombinant TCRs. Perturbing the cellular environment suppressed 2D on-rates but had no effect on 2D off-rate regardless of whether force was applied. In contrast, for the TCR interacting with its monoclonal antibody, the 2D on-rate was insensitive to cellular perturbations and the force-dependent off-rates were indistinguishable for native and recombinant TCRs. These data present novel features of TCR-pMHC kinetics that are regulated by the cellular environment, underscoring the limitations of 3D kinetics in predicting T-cell functions and calling for further elucidation of the underlying molecular and cellular mechanisms that regulate 2D kinetics in physiological settings.
Collapse
Affiliation(s)
- Baoyu Liu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Wei Chen
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kannan Natarajan
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Zhenhai Li
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - David H Margulies
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
36
|
Klotzsch E, Stiegler J, Ben-Ishay E, Gaus K. Do mechanical forces contribute to nanoscale membrane organisation in T cells? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:822-9. [PMID: 25447546 DOI: 10.1016/j.bbamcr.2014.10.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/26/2014] [Accepted: 10/25/2014] [Indexed: 12/23/2022]
Abstract
Mechanotransduction describes how a cell senses and interacts with its environment. The concept originated in adhesion biology where adhesion receptors, integrins, facilitate force transmission between the extracellular matrix and the intracellular actin cytoskeleton. Indeed, during any adhesive contacts, cells do exert mechanical force. Hence, the probing of the local environment by cells results in mechanical cues that contribute to cellular functions and cell fate decisions such as migration, proliferation, differentiation and apoptosis. On the molecular level, mechanical forces can rearrange proteins laterally within the membrane, regulate their activity by inducing conformational changes and probe the mechanical properties and bond strength of receptor-ligands. From this point of view, it appears surprising that molecular forces have been largely overlooked in membrane organisation and ligand discrimination processes in lymphocytes. During T cell activation, the T cell receptor recognises and distinguishes antigenic from benign endogenous peptides to initiate the reorganisation of membrane proteins into signalling clusters within the immunological synapse. In this review, we asked whether characteristics of fibroblast force sensing could be applied to immune cell antigen recognition and signalling, and outline state-of-the-art experimental strategies for studying forces in the context of membrane organisation. This article is part of a Special Issue entitled: Nanoscale membrane orgainisation and signalling.
Collapse
Affiliation(s)
- Enrico Klotzsch
- Centre for Vascular Research, ARC Centre of Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia.
| | - Johannes Stiegler
- Centre for Vascular Research, ARC Centre of Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia
| | - Eldad Ben-Ishay
- Centre for Vascular Research, ARC Centre of Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia
| | - Katharina Gaus
- Centre for Vascular Research, ARC Centre of Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
37
|
Neve-Oz Y, Razvag Y, Sajman J, Sherman E. Mechanisms of localized activation of the T cell antigen receptor inside clusters. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:810-21. [PMID: 25300584 DOI: 10.1016/j.bbamcr.2014.09.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/07/2014] [Accepted: 09/29/2014] [Indexed: 01/21/2023]
Abstract
The T cell antigen receptor (TCR) has been shown to cluster both before and upon engagement with cognate antigens. However, the effect of TCR clustering on its activation remains poorly understood. Here, we used two-color photo-activated localization microscopy (PALM) to visualize individual molecules of TCR and ZAP-70, as a marker of TCR activation and phosphorylation, at the plasma membrane of uniformly activated T cells. Imaging and second-order statistics revealed that ZAP-70 recruitment and TCR activation localized inside TCR clusters. Live cell PALM imaging showed that the extent of localized TCR activation decreased, yet remained significant, with cell spreading. Using dynamic modeling and Monte-Carlo simulations we evaluated possible mechanisms of localized TCR activation. Our simulations indicate that localized TCR activation is the result of long-range cooperative interactions between activated TCRs, or localized activation by Lck and Fyn. Our results demonstrate the role of molecular clustering in cell signaling and activation, and are relevant to studying a wide range of multi-molecular complexes. This article is part of a Special Issue entitled: Nanoscale membrane organisation and signalling.
Collapse
Affiliation(s)
- Yair Neve-Oz
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| | - Yair Razvag
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| | - Julia Sajman
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel.
| |
Collapse
|
38
|
|
39
|
Moore JR. The benefits of diversity: heterogenous DC populations allow for both immunity and tolerance. J Theor Biol 2014; 357:86-102. [PMID: 24816181 DOI: 10.1016/j.jtbi.2014.04.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 04/07/2014] [Accepted: 04/24/2014] [Indexed: 01/31/2023]
Abstract
The immune system must simultaneously mount a response against foreign antigens while tolerating self. How this happens is still unclear as many mechanisms of immune tolerance are antigen non-specific. Antigen specific immune cells called T-cells must first bind to Immunogenic Dendritic Cells (iDCs) before activating and proliferating. These iDCs present both self and foreign antigens during infection, so it is unclear how the immune response can be limited to primarily foreign reactive T-cells. Regulatory T-cells (Tregs) are known to play a key role in self-tolerance. Although they are antigen specific, they also act in an antigen non-specific manner by competing for space and growth factors as well as modifying DC behavior to help kill or deactivate other T-cells. In prior models, the lack of antigen specific control has made simultaneous foreign-immunity and self-tolerance extremely unlikely. We include a heterogeneous DC population, in which different DCs present antigens at different levels. In addition, we include Tolerogenic DC (tDCs) which can delete self-reactive T-cells under normal physiological conditions. We compare different mathematical models of immune tolerance with and without Tregs and heterogenous antigen presentation. For each model, we compute the final number of foreign-reactive and self-reactive T-cells, under a variety of different situations. We find that even if iDCs present more self-antigen than foreign antigen, the immune response will be primarily foreign-reactive as long as there is sufficient presentation of self-antigen on tDCs. Tregs are required primarily for rare or cryptic self-antigens that do not appear frequently on tDCs. We also find that Tregs can only be effective when we include heterogenous antigen presentation, as this allows Tregs and T-cells of the same antigen-specificity to colocalize to the same set of DCs. Tregs better aid immune tolerance when they can both compete for space and growth factors and directly eliminate other T-cells. Our results show the importance of the structure of the DC population in immune tolerance as well as the relative contribution of different cellular mechanisms.
Collapse
Affiliation(s)
- James R Moore
- Department of Mathematics, University of Utah, 155 S 1400 E Rm 233, Salt Lake City, UT 84111, United States.
| |
Collapse
|
40
|
Abstract
T cells are key players of the mammalian adaptive immune system. They experience different mechanical microenvironments during their life cycle, from the thymus, secondary lymph organs, and peripheral tissues that are free of externally applied force, but display variable substrate rigidities to the blood and lymphatic circulation systems, where complicated hydrodynamic forces are present. Regardless of whether T cells are subject to external forces or generate their own internal forces, they respond and adapt to different biomechanical cues to modulate their adhesion, migration, trafficking, and triggering of immune functions through mechanical regulation of various molecules that bear force. These include adhesive receptors, immunoreceptors, motor proteins, cytoskeletal proteins, and their associated molecules. Here, we discuss the forces acting on various surface and cytoplasmic proteins of a T cell in different mechanical milieus. We review existing data on how force regulates protein conformational changes and interactions with counter molecules, including integrins, actin, and the T-cell receptor, and how each relates to T-cell functions.
Collapse
Affiliation(s)
- Wei Chen
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | |
Collapse
|
41
|
Castro M, van Santen HM, Férez M, Alarcón B, Lythe G, Molina-París C. Receptor Pre-Clustering and T cell Responses: Insights into Molecular Mechanisms. Front Immunol 2014; 5:132. [PMID: 24817867 PMCID: PMC4012210 DOI: 10.3389/fimmu.2014.00132] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 03/15/2014] [Indexed: 11/13/2022] Open
Abstract
T cell activation, initiated by T cell receptor (TCR) mediated recognition of pathogen-derived peptides presented by major histocompatibility complex class I or II molecules (pMHC), shows exquisite specificity and sensitivity, even though the TCR-pMHC binding interaction is of low affinity. Recent experimental work suggests that TCR pre-clustering may be a mechanism via which T cells can achieve such high sensitivity. The unresolved stoichiometry of the TCR makes TCR-pMHC binding and TCR triggering, an open question. We formulate a mathematical model to characterize the pre-clustering of T cell receptors (TCRs) on the surface of T cells, motivated by the experimentally observed distribution of TCR clusters on the surface of naive and memory T cells. We extend a recently introduced stochastic criterion to compute the timescales of T cell responses, assuming that ligand-induced cross-linked TCR is the minimum signaling unit. We derive an approximate formula for the mean time to signal initiation. Our results show that pre-clustering reduces the mean activation time. However, additional mechanisms favoring the existence of clusters are required to explain the difference between naive and memory T cell responses. We discuss the biological implications of our results, and both the compatibility and complementarity of our approach with other existing mathematical models.
Collapse
Affiliation(s)
- Mario Castro
- Grupo de Dinámica No-Lineal and Grupo Interdisciplinar de Sistemas Complejos (GISC), Escuela Técnica Superior de Ingeniería (ICAI), Universidad Pontificia Comillas , Madrid , Spain
| | - Hisse M van Santen
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid , Madrid , Spain
| | - María Férez
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid , Madrid , Spain
| | - Balbino Alarcón
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid , Madrid , Spain
| | - Grant Lythe
- Department of Applied Mathematics, School of Mathematics, University of Leeds , Leeds , UK
| | - Carmen Molina-París
- Department of Applied Mathematics, School of Mathematics, University of Leeds , Leeds , UK
| |
Collapse
|
42
|
Dushek O, van der Merwe PA. An induced rebinding model of antigen discrimination. Trends Immunol 2014; 35:153-8. [PMID: 24636916 PMCID: PMC3989030 DOI: 10.1016/j.it.2014.02.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 02/10/2014] [Accepted: 02/11/2014] [Indexed: 12/01/2022]
Abstract
We propose that pMHC binding to TCR can increase (induce) pMHC rebinding. Published studies are consistent with an induced rebinding model. Induced rebinding improves the ability of T cells to discriminate antigens. Induced rebinding relates 3D to 2D TCR–pMHC binding parameters.
T cells have to detect rare high-affinity ‘foreign’ peptide MHC (pMHC) ligands among abundant low-affinity ‘self’-peptide MHC ligands. It remains unclear how this remarkable discrimination is achieved. Kinetic proofreading mechanisms can provide the required specificity but only at the expense of much reduced sensitivity. A number of recent observations suggest that pMHC engagement of T cell receptors (TCRs) induces changes such as clustering and/or conformational alterations that enhance subsequent rebinding. We show that inclusion of induced rebinding to the same pMHC in kinetic proofreading models enhances the sensitivity of TCR recognition while retaining specificity. Moreover, induced rebinding is able to reproduce the striking, and hitherto unexplained, 2D membrane-binding properties recently reported for the TCR.
Collapse
Affiliation(s)
- Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK; Wolfson Centre for Mathematical Biology, University of Oxford, Oxford, UK.
| | | |
Collapse
|
43
|
|
44
|
Bolel P, Datta S, Mahapatra N, Halder M. Exploration of pH-Dependent Behavior of the Anion Receptor Pocket of Subdomain IIA of HSA: Determination of Effective Pocket Charge Using the Debye–Hückel Limiting Law. J Phys Chem B 2013; 118:26-36. [DOI: 10.1021/jp407057f] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Priyanka Bolel
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
| | - Shubhashis Datta
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
| | - Niharendu Mahapatra
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
| | - Mintu Halder
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
| |
Collapse
|
45
|
Improved ligand discrimination by force-induced unbinding of the T cell receptor from peptide-MHC. Biophys J 2013; 104:1670-5. [PMID: 23601314 DOI: 10.1016/j.bpj.2013.03.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 03/11/2013] [Accepted: 03/15/2013] [Indexed: 01/22/2023] Open
Abstract
T cell activation is mediated via the recognition of peptides by the T cell receptor (TCR). This receptor ligand interaction is highly specific, and the TCR has to discriminate between a huge number of peptides presented by the products of the major histocompatibility complexes (MHCs). Recent studies indicate that cells probe the TCR-pMHC interaction by imposing force on the interaction. Here we investigated in a theoretical analysis the consequences of such force-induced unbinding for T cell recognition. Our findings are as follows. First, the bond rupture under force is much faster, improving the time resolution of the discrimination process. Second, cells can access additional parameters characterizing the shape of the binding energy surface. Third, load-induced unbinding yields a reduced coefficient of variation of the bond lifetimes, which improves the discriminative power even between peptide/MHCs (pMHCs) with similar off-rates.
Collapse
|
46
|
Míguez DG, Gil-Guiñón E, Pons S, Martí E. Smad2 and Smad3 cooperate and antagonize simultaneously in vertebrate neurogenesis. J Cell Sci 2013; 126:5335-43. [DOI: 10.1242/jcs.130435] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The transforming growth factor beta (TGF-β) pathway plays key roles in development and cancer. (TGF-β) signaling converges on the Smad2 and Smad3 effectors, which can either cooperate or antagonize to regulate their transcriptional targets. Here we performed in vivo and in silico experiments to study how such cooperativity and antagonism might function during neurogenesis. In vivo electroporation experiments in the chick embryo neural tube show that Smad2 and Smad3 cooperate to promote neurogenesis, as well as the transcription of Smad3 specific targets. Smad2 knockdown enhances neurogenesis and the transcription of Smad3 specific targets. A mathematical model of the TGF-β pathway fits the experimental results and predicts that the proportions of the three different trimeric complexes formed dictates the transcriptional responses of the R-Smads. As such, Smad2 targets are activated solely by the Smad2-Smad2-Smad4 complex, while Smad3 targets are activated both by Smad2-Smad3 Smad4 and Smad3- Smad3-Smad4 trimers. Since we have modeled the Smad responses onto arbitrary genes, we propose that this mechanism might be extended to additional activities of TGF-β in development and disease.
Collapse
|
47
|
Abstract
Leukocytes play a critical role in recognizing and responding to infection and cancer. Central to this function is an array of cell-surface receptors that lack sequence homology. Many of these receptors have in common the fact that their signaling involves phosphorylation of cytoplasmic domains by extrinsic tyrosine kinases. These non-catalytic tyrosine-phosphorylated receptors (NTRs) share a number of other features, including small size and optimal stimulation by surface-associated ligands. We argue here that NTRs are also likely to share the same kinetic-segregation triggering mechanism, which involves segregation of the engaged NTR from receptor tyrosine phosphatases with large ectodomains such as CD45 and CD148. NTRs signal through tyrosine-containing cytoplasmic motifs, which recruit distinct cytoplasmic signaling proteins when phosphorylated, transducing activatory or inhibitory signals. They have two features that make them uniquely well suited to their role in immune recognition of infection and cancer. Their modular structure enables the coupling of many rapidly evolving receptors with diverse ligand specificities to the same conserved signaling machinery. Their similarity in size and shared signaling machinery enables them to colocalize at cell-cell interfaces when they engage ligands, facilitating the integration of activatory and inhibitory signals from multiple receptors at the cell surface.
Collapse
Affiliation(s)
- Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, UK
| | | | | |
Collapse
|
48
|
Zarnitsyna V, Zhu C. T cell triggering: insights from 2D kinetics analysis of molecular interactions. Phys Biol 2012; 9:045005. [PMID: 22871794 DOI: 10.1088/1478-3975/9/4/045005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Interaction of the T cell receptor (TCR) with pathogen-derived peptide presented by the major histocompatibility complex (pMHC) molecule is central to adaptive immunity as it initiates intracellular signaling to trigger T cell response to infection. Kinetic parameters of this interaction have been under intensive investigation for more than two decades using soluble pMHCs and/or TCRs with at least one of them in the solution (three-dimensional (3D) methods). Recently, several techniques have been developed to enable kinetic analysis on live T cells with pMHCs presented by surrogate antigen presenting cells (APCs) or supported planar lipid bilayers (two-dimensional (2D) methods). Comparison of 2D versus 3D parameters reveals drastic differences with broader ranges of 2D affinities and on-rates and orders of magnitude faster 2D off-rates for functionally distinct pMHCs. Here we review new 2D data and discuss how it may impact previously developed models of T cell discrimination between pMHCs of different potencies.
Collapse
Affiliation(s)
- Veronika Zarnitsyna
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA.
| | | |
Collapse
|
49
|
Ma Z, Discher DE, Finkel TH. Mechanical force in T cell receptor signal initiation. Front Immunol 2012; 3:217. [PMID: 22833746 PMCID: PMC3400889 DOI: 10.3389/fimmu.2012.00217] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 07/04/2012] [Indexed: 11/15/2022] Open
Affiliation(s)
- Zhengyu Ma
- Department of Pediatrics, Nemours Children's Hospital Orlando, FL, USA
| | | | | |
Collapse
|