101
|
Kervevan J, Chakrabarti LA. Role of CD4+ T Cells in the Control of Viral Infections: Recent Advances and Open Questions. Int J Mol Sci 2021; 22:E523. [PMID: 33430234 PMCID: PMC7825705 DOI: 10.3390/ijms22020523] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/26/2022] Open
Abstract
CD4+ T cells orchestrate adaptive immune responses through their capacity to recruit and provide help to multiple immune effectors, in addition to exerting direct effector functions. CD4+ T cells are increasingly recognized as playing an essential role in the control of chronic viral infections. In this review, we present recent advances in understanding the nature of CD4+ T cell help provided to antiviral effectors. Drawing from our studies of natural human immunodeficiency virus (HIV) control, we then focus on the role of high-affinity T cell receptor (TCR) clonotypes in mediating antiviral CD4+ T cell responses. Last, we discuss the role of TCR affinity in determining CD4+ T cell differentiation, reviewing the at times divergent studies associating TCR signal strength to the choice of a T helper 1 (Th1) or a T follicular helper (Tfh) cell fate.
Collapse
Affiliation(s)
- Jérôme Kervevan
- Control of Chronic Viral Infections Group (CIVIC), Virus and Immunity Unit, Institut Pasteur, 75724 Paris, France;
- CNRS UMR, 3569 Paris, France
| | - Lisa A. Chakrabarti
- Control of Chronic Viral Infections Group (CIVIC), Virus and Immunity Unit, Institut Pasteur, 75724 Paris, France;
- CNRS UMR, 3569 Paris, France
| |
Collapse
|
102
|
TCR Recognition of Peptide-MHC-I: Rule Makers and Breakers. Int J Mol Sci 2020; 22:ijms22010068. [PMID: 33374673 PMCID: PMC7793522 DOI: 10.3390/ijms22010068] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
T cells are a critical part of the adaptive immune system that are able to distinguish between healthy and unhealthy cells. Upon recognition of protein fragments (peptides), activated T cells will contribute to the immune response and help clear infection. The major histocompatibility complex (MHC) molecules, or human leukocyte antigens (HLA) in humans, bind these peptides to present them to T cells that recognise them with their surface T cell receptors (TCR). This recognition event is the first step that leads to T cell activation, and in turn can dictate disease outcomes. The visualisation of TCR interaction with pMHC using structural biology has been crucial in understanding this key event, unravelling the parameters that drive this interaction and their impact on the immune response. The last five years has been the most productive within the field, wherein half of current unique TCR-pMHC-I structures to date were determined within this time. Here, we review the new insights learned from these recent TCR-pMHC-I structures and their impact on T cell activation.
Collapse
|
103
|
Regulations of T Cell Activation by Membrane and Cytoskeleton. MEMBRANES 2020; 10:membranes10120443. [PMID: 33352750 PMCID: PMC7765812 DOI: 10.3390/membranes10120443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022]
Abstract
Among various types of membrane proteins that are regulated by cytoskeleton, the T cell receptor (TCR) greatly benefits from these cellular machineries for its function. The T cell is activated by the ligation of TCR to its target agonist peptide. However, the binding affinity of the two is not very strong, while the T cell needs to discriminate agonist from many nonagonist peptides. Moreover, the strength and duration of the activation signaling need to be tuned for immunological functions. Many years of investigations revealed that dynamic acto-myosin cytoskeletons and plasma membranes in T cells facilitate such regulations by modulating the spatiotemporal distributions of proteins in plasma membranes and by applying mechanical loads on proteins. In these processes, protein dynamics in multiple scales are involved, ranging from collective molecular motions and macroscopic molecular organizations at the cell–cell interface to microscopic changes in distances between receptor and ligand molecules. In this review, details of how cytoskeletons and membranes regulate these processes are discussed, with the emphasis on how all these processes are coordinated to occur within a single cell system.
Collapse
|
104
|
TCR Transgenic Mice: A Valuable Tool for Studying Viral Immunopathogenesis Mechanisms. Int J Mol Sci 2020; 21:ijms21249690. [PMID: 33353154 PMCID: PMC7765986 DOI: 10.3390/ijms21249690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 01/07/2023] Open
Abstract
Viral infectious diseases are a significant burden on public health and the global economy, and new viral threats emerge continuously. Since CD4+ and CD8+ T cell responses are essential to eliminating viruses, it is important to understand the underlying mechanisms of anti-viral T cell-mediated immunopathogenesis during viral infections. Remarkable progress in transgenic (Tg) techniques has enabled scientists to more readily understand the mechanisms of viral pathogenesis. T cell receptor (TCR) Tg mice are extremely useful in studying T cell-mediated immune responses because the majority of T cells in these mice express specific TCRs for partner antigens. In this review, we discuss the important studies utilizing TCR Tg mice to unveil underlying mechanisms of T cell-mediated immunopathogenesis during viral infections.
Collapse
|
105
|
Li R, Ma C, Cai H, Chen W. The CAR T-Cell Mechanoimmunology at a Glance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002628. [PMID: 33344135 PMCID: PMC7740088 DOI: 10.1002/advs.202002628] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/13/2020] [Indexed: 05/10/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell transfer is a novel paradigm of adoptive T-cell immunotherapy. When coming into contact with a target cancer cell, CAR T-cell forms a nonclassical immunological synapse with the cancer cell and dynamically orchestrates multiple critical forces to commit cytotoxic immune function. Such an immunologic process involves a force transmission in the CAR and a spatiotemporal remodeling of cell cytoskeleton to facilitate CAR activation and CAR T-cell cytotoxic function. Yet, the detailed understanding of such mechanotransduction at the interface between the CAR T-cell and the target cell, as well as its molecular structure and signaling, remains less defined and is just beginning to emerge. This article summarizes the basic mechanisms and principles of CAR T-cell mechanoimmunology, and various lessons that can be comparatively learned from interrogation of mechanotransduction at the immunological synapse in normal cytotoxic T-cell. The recent development and future application of novel bioengineering tools for studying CAR T-cell mechanoimmunology is also discussed. It is believed that this progress report will shed light on the CAR T-cell mechanoimmunology and encourage future researches in revealing the less explored yet important mechanosensing and mechanotransductive mechanisms involved in CAR T-cell immuno-oncology.
Collapse
Affiliation(s)
- Rui Li
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNY11201USA
- Department of Biomedical EngineeringNew York UniversityBrooklynNY11201USA
| | - Chao Ma
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNY11201USA
- Department of Biomedical EngineeringNew York UniversityBrooklynNY11201USA
| | - Haogang Cai
- Tech4Health instituteNYU Langone HealthNew YorkNY10016USA
- Department of RadiologyNYU Langone HealthNew YorkNY10016USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNY11201USA
- Department of Biomedical EngineeringNew York UniversityBrooklynNY11201USA
- Laura and Isaac Perlmutter Cancer CenterNYU Langone HealthNew YorkNY10016USA
| |
Collapse
|
106
|
Kolawole EM, Lamb TJ, Evavold BD. Relationship of 2D Affinity to T Cell Functional Outcomes. Int J Mol Sci 2020; 21:E7969. [PMID: 33120989 PMCID: PMC7662510 DOI: 10.3390/ijms21217969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022] Open
Abstract
T cells are critical for a functioning adaptive immune response and a strong correlation exists between T cell responses and T cell receptor (TCR): peptide-loaded MHC (pMHC) binding. Studies that utilize pMHC tetramer, multimers, and assays of three-dimensional (3D) affinity have provided advancements in our understanding of T cell responses across different diseases. However, these technologies focus on higher affinity and avidity T cells while missing the lower affinity responders. Lower affinity TCRs in expanded polyclonal populations almost always constitute a significant proportion of the response with cells mediating different effector functions associated with variation in the proportion of high and low affinity T cells. Since lower affinity T cells expand and are functional, a fully inclusive view of T cell responses is required to accurately interpret the role of affinity for adaptive T cell immunity. For example, low affinity T cells are capable of inducing autoimmune disease and T cells with an intermediate affinity have been shown to exhibit an optimal anti-tumor response. Here, we focus on how affinity of the TCR may relate to T cell phenotype and provide examples where 2D affinity influences functional outcomes.
Collapse
Affiliation(s)
| | | | - Brian D. Evavold
- Department of Pathology, University of Utah, 15 N Medical Drive, Salt Lake City, UT 84112, USA; (E.M.K.); (T.J.L.)
| |
Collapse
|
107
|
Zhang X, Mariano CF, Ando Y, Shen K. Bioengineering tools for probing intracellular events in T lymphocytes. WIREs Mech Dis 2020; 13:e1510. [PMID: 33073545 DOI: 10.1002/wsbm.1510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 11/11/2022]
Abstract
T lymphocytes are the central coordinator and executor of many immune functions. The activation and function of T lymphocytes are mediated through the engagement of cell surface receptors and regulated by a myriad of intracellular signaling network. Bioengineering tools, including imaging modalities and fluorescent probes, have been developed and employed to elucidate the cellular events throughout the functional lifespan of T cells. A better understanding of these events can broaden our knowledge in the immune systems biology, as well as accelerate the development of effective diagnostics and immunotherapies. Here we review the commonly used and recently developed techniques and probes for monitoring T lymphocyte intracellular events, following the order of intracellular events in T cells from activation, signaling, metabolism to apoptosis. The techniques introduced here can be broadly applied to other immune cells and cell systems. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology Immune System Diseases > Biomedical Engineering Infectious Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Chelsea F Mariano
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA.,USC Stem Cell, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
108
|
Singh NK, Alonso JA, Harris DT, Anderson SD, Ma J, Hellman LM, Rosenberg AM, Kolawole EM, Evavold BD, Kranz DM, Baker BM. An Engineered T Cell Receptor Variant Realizes the Limits of Functional Binding Modes. Biochemistry 2020; 59:4163-4175. [PMID: 33074657 DOI: 10.1021/acs.biochem.0c00689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
T cell receptors (TCRs) orchestrate cellular immunity by recognizing peptides presented by a range of major histocompatibility complex (MHC) proteins. Naturally occurring TCRs bind the composite peptide/MHC surface, recognizing peptides that are structurally and chemically compatible with the TCR binding site. Here we describe a molecularly evolved TCR variant that binds the human class I MHC protein HLA-A2 independent of the bound peptide, achieved by a drastic perturbation of the TCR binding geometry that places the molecule far from the peptide binding groove. This unique geometry is unsupportive of normal T cell signaling. A substantial divergence between affinity measurements in solution and in two dimensions between proximal cell membranes leads us to attribute the lack of signaling to steric hindrance that limits binding in the confines of a cell-cell interface. Our results provide an example of how receptor binding geometry can impact T cell function and provide further support for the view that germline-encoded residues in TCR binding loops evolved to drive productive TCR recognition and signaling.
Collapse
Affiliation(s)
- Nishant K Singh
- Department of Chemistry and Biochemistry and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Jesus A Alonso
- Department of Chemistry and Biochemistry and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Daniel T Harris
- Department of Biochemistry and Cancer Center at Illinois, University of Illinois, Urbana, Illinois 61801, United States
| | - Scott D Anderson
- Department of Biochemistry and Cancer Center at Illinois, University of Illinois, Urbana, Illinois 61801, United States
| | - Jiaqi Ma
- Department of Chemistry and Biochemistry and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Lance M Hellman
- Department of Chemistry and Biochemistry and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Aaron M Rosenberg
- Department of Chemistry and Biochemistry and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Elizabeth M Kolawole
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Brian D Evavold
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah 84112, United States
| | - David M Kranz
- Department of Biochemistry and Cancer Center at Illinois, University of Illinois, Urbana, Illinois 61801, United States
| | - Brian M Baker
- Department of Chemistry and Biochemistry and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
109
|
Chandler NJ, Call MJ, Call ME. T Cell Activation Machinery: Form and Function in Natural and Engineered Immune Receptors. Int J Mol Sci 2020; 21:E7424. [PMID: 33050044 PMCID: PMC7582382 DOI: 10.3390/ijms21197424] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
The impressive success of chimeric antigen receptor (CAR)-T cell therapies in treating advanced B-cell malignancies has spurred a frenzy of activity aimed at developing CAR-T therapies for other cancers, particularly solid tumors, and optimizing engineered T cells for maximum clinical benefit in many different disease contexts. A rapidly growing body of design work is examining every modular component of traditional single-chain CARs as well as expanding out into many new and innovative engineered immunoreceptor designs that depart from this template. New approaches to immune cell and receptor engineering are being reported with rapidly increasing frequency, and many recent high-quality reviews (including one in this special issue) provide comprehensive coverage of the history and current state of the art in CAR-T and related cellular immunotherapies. In this review, we step back to examine our current understanding of the structure-function relationships in natural and engineered lymphocyte-activating receptors, with an eye towards evaluating how well the current-generation CAR designs recapitulate the most desirable features of their natural counterparts. We identify key areas that we believe are under-studied and therefore represent opportunities to further improve our grasp of form and function in natural and engineered receptors and to rationally design better therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Chandler
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Melissa J. Call
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Matthew E. Call
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
110
|
How the T cell signaling network processes information to discriminate between self and agonist ligands. Proc Natl Acad Sci U S A 2020; 117:26020-26030. [PMID: 33020303 DOI: 10.1073/pnas.2008303117] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
T cells exhibit remarkable sensitivity and selectivity in detecting and responding to agonist peptides (p) bound to MHC molecules in a sea of self pMHC molecules. Despite much work, understanding of the underlying mechanisms of distinguishing such ligands remains incomplete. Here, we quantify T cell discriminatory capacity using channel capacity, a direct measure of the signaling network's ability to discriminate between antigen-presenting cells (APCs) displaying either self ligands or a mixture of self and agonist ligands. This metric shows how differences in information content between these two types of peptidomes are decoded by the topology and rates of kinetic proofreading signaling steps inside T cells. Using channel capacity, we constructed numerically substantiated hypotheses to explain the discriminatory role of a recently identified slow LAT Y132 phosphorylation step. Our results revealed that in addition to the number and kinetics of sequential signaling steps, a key determinant of discriminatory capability is spatial localization of a minimum number of these steps to the engaged TCR. Biochemical and imaging experiments support these findings. Our results also reveal the discriminatory role of early negative feedback and necessary amplification conferred by late positive feedback.
Collapse
|
111
|
Sung CL, Hung Y, Rittase W, Zhu C, Wu CFJ. Calibration for Computer Experiments With Binary Responses and Application to Cell Adhesion Study. J Am Stat Assoc 2020. [DOI: 10.1080/01621459.2019.1699419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Chih-Li Sung
- Department of Statistics and Probability, Michigan State University, East Lansing, MI
| | - Ying Hung
- Department of Statistics, Rutgers, The State University of New Jersey, Piscataway, NJ
| | - William Rittase
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Cheng Zhu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - C. F. J. Wu
- School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
112
|
Junghans V, Chouliara M, Santos AM, Hatherley D, Petersen J, Dam T, Svensson LM, Rossjohn J, Davis SJ, Jönsson P. Effects of a local auxiliary protein on the two-dimensional affinity of a TCR-peptide MHC interaction. J Cell Sci 2020; 133:jcs245985. [PMID: 32591485 DOI: 10.1242/jcs.245985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022] Open
Abstract
The affinity of T-cell receptors (TCRs) for major histocompatibility complex molecules (MHCs) presenting cognate antigens likely determines whether T cells initiate immune responses, or not. There exist few measurements of two-dimensional (2D) TCR-MHC interactions, and the effect of auxiliary proteins on binding is unexplored. Here, Jurkat T-cells expressing the MHC molecule HLA-DQ8-glia-α1 and the ligand of an adhesion protein (rat CD2) were allowed to bind supported lipid bilayers (SLBs) presenting fluorescently labelled L3-12 TCR and rat CD2, allowing measurements of binding unconfounded by cell signaling effects or co-receptor binding. The 2D Kd for L3-12 TCR binding to HLA-DQ8-glia-α1, of 14±5 molecules/μm2 (mean±s.d.), was only marginally influenced by including CD2 up to ∼200 bound molecules/μm2 but higher CD2 densities reduced the affinity up to 1.9-fold. Cell-SLB contact size increased steadily with ligand density without affecting binding for contacts at up to ∼20% of total cell area, but beyond this lamellipodia appeared, giving an apparent increase in bound receptors of up to 50%. Our findings show how parameters other than the specific protein-protein interaction can influence binding behavior at cell-cell contacts.
Collapse
Affiliation(s)
| | - Manto Chouliara
- Department of Chemistry, Lund University, 221 00 Lund, Sweden
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford; and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, UK
| | - Deborah Hatherley
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford; and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, UK
| | - Jan Petersen
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Tommy Dam
- Department of Chemistry, Lund University, 221 00 Lund, Sweden
| | - Lena M Svensson
- Department of Experimental Medical Science, Lund University, 221 00 Lund, Sweden; and School of Medical Sciences, Örebro University, 701 82 Örebro, Sweden
| | - Jamie Rossjohn
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford; and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, UK
| | - Peter Jönsson
- Department of Chemistry, Lund University, 221 00 Lund, Sweden
| |
Collapse
|
113
|
An C, Hu W, Gao J, Ju BF, Obeidy P, Zhao YC, Tu X, Fang W, Ju LA, Chen W. Ultra-stable Biomembrane Force Probe for Accurately Determining Slow Dissociation Kinetics of PD-1 Blockade Antibodies on Single Living Cells. NANO LETTERS 2020; 20:5133-5140. [PMID: 32530632 DOI: 10.1021/acs.nanolett.0c01360] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Immune checkpoint blockade with monoclonal antibodies (mAbs) that target programmed cell death protein-1 (PD-1) has remarkably revolutionized cancer therapy. Their binding kinetics measured by surface plasmon resonance does not always correlate well with their immunotherapeutic efficacies, mainly due to the lack of two-dimensional cell plasma membrane and the capability of force sensing and manipulation. In this regard, based on a more suitable and ultra-sensitive biomechanical nanotool, biomembrane force probe (BFP), we developed a Double-edge Smart Feedback control system as an ultra-stable platform to characterize ultra-long bond lifetimes of receptor-ligand binding on living cells. We further benchmarked the dissociation kinetics for three clinically approved PD-1 blockade mAbs (Nivolumab, Pembrolizumab, and Camrelizumab), intriguingly correlating well with the objective response rates in the hepatocellular carcinoma second-line treatment. This ultra-stable BFP potentially provides a compelling kinetic platform to direct the screening, optimization, and clinical selection of therapeutic antibodies in the future.
Collapse
Affiliation(s)
- Chenyi An
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Fluid Power and Mechatronic Systems, Key Laboratory for Biomedical Engineering of Ministry of Education, and School of Mechanical Engineering, Zhejiang University, Hangzhou, China, 310058
| | - Wei Hu
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Fluid Power and Mechatronic Systems, Key Laboratory for Biomedical Engineering of Ministry of Education, and School of Mechanical Engineering, Zhejiang University, Hangzhou, China, 310058
| | - Jie Gao
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Fluid Power and Mechatronic Systems, Key Laboratory for Biomedical Engineering of Ministry of Education, and School of Mechanical Engineering, Zhejiang University, Hangzhou, China, 310058
| | - Bing-Feng Ju
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Fluid Power and Mechatronic Systems, Key Laboratory for Biomedical Engineering of Ministry of Education, and School of Mechanical Engineering, Zhejiang University, Hangzhou, China, 310058
| | - Peyman Obeidy
- School of Biomedical Engineering, Faculty of Engineering and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales Australia, 2006
| | - Yunduo Charles Zhao
- School of Biomedical Engineering, Faculty of Engineering and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales Australia, 2006
| | - Xiaoxuan Tu
- Department of Medical Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 310000
| | - Weijia Fang
- Department of Medical Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 310000
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 310000
| | - Lining Arnold Ju
- School of Biomedical Engineering, Faculty of Engineering and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales Australia, 2006
- Heart Research Institute, Newtown, New South Wales Australia, 2042
| | - Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Fluid Power and Mechatronic Systems, Key Laboratory for Biomedical Engineering of Ministry of Education, and School of Mechanical Engineering, Zhejiang University, Hangzhou, China, 310058
- State Key Laboratory for Modern Optical Instrumentation and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China, 310058
| |
Collapse
|
114
|
Klotz L, Eschborn M, Lindner M, Liebmann M, Herold M, Janoschka C, Torres Garrido B, Schulte-Mecklenbeck A, Gross CC, Breuer J, Hundehege P, Posevitz V, Pignolet B, Nebel G, Glander S, Freise N, Austermann J, Wirth T, Campbell GR, Schneider-Hohendorf T, Eveslage M, Brassat D, Schwab N, Loser K, Roth J, Busch KB, Stoll M, Mahad DJ, Meuth SG, Turner T, Bar-Or A, Wiendl H. Teriflunomide treatment for multiple sclerosis modulates T cell mitochondrial respiration with affinity-dependent effects. Sci Transl Med 2020; 11:11/490/eaao5563. [PMID: 31043571 DOI: 10.1126/scitranslmed.aao5563] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 08/20/2018] [Accepted: 04/02/2019] [Indexed: 01/06/2023]
Abstract
Interference with immune cell proliferation represents a successful treatment strategy in T cell-mediated autoimmune diseases such as rheumatoid arthritis and multiple sclerosis (MS). One prominent example is pharmacological inhibition of dihydroorotate dehydrogenase (DHODH), which mediates de novo pyrimidine synthesis in actively proliferating T and B lymphocytes. Within the TERIDYNAMIC clinical study, we observed that the DHODH inhibitor teriflunomide caused selective changes in T cell subset composition and T cell receptor repertoire diversity in patients with relapsing-remitting MS (RRMS). In a preclinical antigen-specific setup, DHODH inhibition preferentially suppressed the proliferation of high-affinity T cells. Mechanistically, DHODH inhibition interferes with oxidative phosphorylation (OXPHOS) and aerobic glycolysis in activated T cells via functional inhibition of complex III of the respiratory chain. The affinity-dependent effects of DHODH inhibition were closely linked to differences in T cell metabolism. High-affinity T cells preferentially use OXPHOS during early activation, which explains their increased susceptibility toward DHODH inhibition. In a mouse model of MS, DHODH inhibitory treatment resulted in preferential inhibition of high-affinity autoreactive T cell clones. Compared to T cells from healthy controls, T cells from patients with RRMS exhibited increased OXPHOS and glycolysis, which were reduced with teriflunomide treatment. Together, these data point to a mechanism of action where DHODH inhibition corrects metabolic disturbances in T cells, which primarily affects profoundly metabolically active high-affinity T cell clones. Hence, DHODH inhibition may promote recovery of an altered T cell receptor repertoire in autoimmunity.
Collapse
Affiliation(s)
- Luisa Klotz
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany.
| | - Melanie Eschborn
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Maren Lindner
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Marie Liebmann
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Martin Herold
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Claudia Janoschka
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Belén Torres Garrido
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Andreas Schulte-Mecklenbeck
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Catharina C Gross
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Johanna Breuer
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Petra Hundehege
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Vilmos Posevitz
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Béatrice Pignolet
- CRC-SEP, Neurosciences Department, Toulouse University Hospital and INSERM U1043 - CNRS UMR 5282, Centre de Physiopathologie Toulouse-Purpan, Université Toulouse III, 31300 Toulouse, France
| | - Giulia Nebel
- University of Münster, Institute of Molecular Cell Biology, 48149 Münster, Germany
| | - Shirin Glander
- University of Münster, Department of Genetic Epidemiology, 48149 Münster, Germany
| | - Nicole Freise
- University of Münster, Department of Immunology, 48149 Münster, Germany
| | - Judith Austermann
- University of Münster, Department of Immunology, 48149 Münster, Germany
| | - Timo Wirth
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Graham R Campbell
- University of Edinburgh, Centre for Clinical Brain Sciences, EH8 9YL Edinburgh, UK
| | - Tilman Schneider-Hohendorf
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Maria Eveslage
- University of Münster, Institute of Biostatistics and Clinical Research, 48149 Münster, Germany
| | - David Brassat
- CRC-SEP, Neurosciences Department, Toulouse University Hospital and INSERM U1043 - CNRS UMR 5282, Centre de Physiopathologie Toulouse-Purpan, Université Toulouse III, 31300 Toulouse, France
| | - Nicholas Schwab
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | - Karin Loser
- University Hospital Münster, Department of Dermatology, 48149 Münster, Germany
| | - Johannes Roth
- University of Münster, Department of Immunology, 48149 Münster, Germany
| | - Karin B Busch
- University of Münster, Institute of Molecular Cell Biology, 48149 Münster, Germany
| | - Monika Stoll
- University of Münster, Department of Genetic Epidemiology, 48149 Münster, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, Netherlands
| | - Don J Mahad
- University of Edinburgh, Centre for Clinical Brain Sciences, EH8 9YL Edinburgh, UK
| | - Sven G Meuth
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany
| | | | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Heinz Wiendl
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, 48149 Münster, Germany.,Brain and Mind Centre, Medical Faculty, University of Sydney, Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
115
|
Lim Y, Kim S, Kim S, Kim DI, Kang KW, Hong SH, Lee SM, Koh HR, Seo YJ. n-3 Polyunsaturated Fatty Acids Impede the TCR Mobility and the TCR-pMHC Interaction of Anti-Viral CD8+ T Cells. Viruses 2020; 12:v12060639. [PMID: 32545480 PMCID: PMC7354506 DOI: 10.3390/v12060639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/27/2022] Open
Abstract
The immune-suppressive effects of omega-3 (n-3) polyunsaturated fatty acids (PUFAs) on T cells have been observed via multiple in vitro and in vivo models. However, the precise mechanism that causes these effects is still undefined. In this study, we investigated whether n-3 PUFAs regulated T cell receptor (TCR) and peptide-major histocompatibility complex (pMHC) interactions. The expansion of anti-viral CD8+ T cells that endogenously synthesize n-3 PUFAs (FAT-1) dramatically decreased upon lymphocytic choriomeningitis virus (LCMV) infection in vivo. This decrease was not caused by the considerable reduction of TCR expression or the impaired chemotactic activity of T cells. Interestingly, a highly inclined and laminated optical sheet (HILO) microscopic analysis revealed that the TCR motility was notably reduced on the surface of the FAT-1 CD8+ T cells compared to the wild type (WT) CD8+ T cells. Importantly, the adhesion strength of the FAT-1 CD8+ T cells to the peptide-MHC was significantly lower than that of the WT CD8+T cells. Consistent with this result, treatment with docosahexaenoic acid (DHA), one type of n-3 PUFA, significantly decreased CD8+ T cell adhesion to the pMHC. Collectively, our results reveal a novel mechanism through which n-3 PUFAs decrease TCR-pMHC interactions by modulating TCR mobility on CD8+ T cell surfaces.
Collapse
Affiliation(s)
- Younghyun Lim
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea; (Y.L.); (S.K.); (D.-I.K.)
| | - Seyoung Kim
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea; (Y.L.); (S.K.); (D.-I.K.)
| | - Sehoon Kim
- Department of Chemistry, Chung-Ang University, Seoul 06974, Korea;
| | - Dong-In Kim
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea; (Y.L.); (S.K.); (D.-I.K.)
| | - Kyung Won Kang
- Division of Biotechnology, College of Environmental and Bioresources, Jeonbuk National University, Iksan 54596, Korea; (K.W.K.); (S.-M.L.)
| | - So-Hee Hong
- Department of Biotechnology, the Catholic University of Korea, Bucheon 14662, Korea;
| | - Sang-Myeong Lee
- Division of Biotechnology, College of Environmental and Bioresources, Jeonbuk National University, Iksan 54596, Korea; (K.W.K.); (S.-M.L.)
| | - Hye Ran Koh
- Department of Chemistry, Chung-Ang University, Seoul 06974, Korea;
- Correspondence: (H.R.K.); (Y.-J.S.)
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea; (Y.L.); (S.K.); (D.-I.K.)
- Correspondence: (H.R.K.); (Y.-J.S.)
| |
Collapse
|
116
|
Llorente García I, Marsh M. A biophysical perspective on receptor-mediated virus entry with a focus on HIV. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183158. [PMID: 31863725 PMCID: PMC7156917 DOI: 10.1016/j.bbamem.2019.183158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/14/2022]
Abstract
As part of their entry and infection strategy, viruses interact with specific receptor molecules expressed on the surface of target cells. The efficiency and kinetics of the virus-receptor interactions required for a virus to productively infect a cell is determined by the biophysical properties of the receptors, which are in turn influenced by the receptors' plasma membrane (PM) environments. Currently, little is known about the biophysical properties of these receptor molecules or their engagement during virus binding and entry. Here we review virus-receptor interactions focusing on the human immunodeficiency virus type 1 (HIV), the etiological agent of acquired immunodeficiency syndrome (AIDS), as a model system. HIV is one of the best characterised enveloped viruses, with the identity, roles and structure of the key molecules required for infection well established. We review current knowledge of receptor-mediated HIV entry, addressing the properties of the HIV cell-surface receptors, the techniques used to measure these properties, and the macromolecular interactions and events required for virus entry. We discuss some of the key biophysical principles underlying receptor-mediated virus entry and attempt to interpret the available data in the context of biophysical mechanisms. We also highlight crucial outstanding questions and consider how new tools might be applied to advance understanding of the biophysical properties of viral receptors and the dynamic events leading to virus entry.
Collapse
Affiliation(s)
| | - Mark Marsh
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
117
|
Sung CL, Hung Y, Rittase W, Zhu C, Jeff Wu CF. A Generalized Gaussian Process Model for Computer Experiments With Binary Time Series. J Am Stat Assoc 2020. [DOI: 10.1080/01621459.2019.1604361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Chih-Li Sung
- Department of Statistics and Probability, Michigan State University, East Lansing, MI
| | - Ying Hung
- Department of Statistics, Rutgers, The State University of New Jersey, New Brunswick, NJ
| | - William Rittase
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Cheng Zhu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - C. F. Jeff Wu
- School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
118
|
Affiliation(s)
- Pirooz Zareie
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Carine Farenc
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Nicole L. La Gruta
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| |
Collapse
|
119
|
Bettini M, Scavuzzo MA, Liu B, Kolawole E, Guo L, Evavold BD, Borowiak M, Bettini ML. A Critical Insulin TCR Contact Residue Selects High-Affinity and Pathogenic Insulin-Specific T Cells. Diabetes 2020; 69:392-400. [PMID: 31836691 PMCID: PMC7034183 DOI: 10.2337/db19-0821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/08/2019] [Indexed: 02/05/2023]
Abstract
Type 1 diabetes is an autoimmune-mediated disease that culminates in the targeted destruction of insulin-producing β-cells. CD4 responses in NOD mice are dominated by insulin epitope B:9-23 (InsB9-23) specificity, and mutation of the key T-cell receptor (TCR) contact residue within the epitope prevents diabetes development. However, it is not clear how insulin self-antigen controls the selection of autoimmune and regulatory T cells (Tregs). Here we demonstrate that mutation of insulin epitope results in escape of highly pathogenic T cells. We observe an increase in antigen reactivity, clonality, and pathogenicity of insulin-specific T cells that develop in the absence of cognate antigen. Using a single TCR system, we demonstrate that Treg development is greatly diminished in mice with the Y16A mutant epitope. Collectively, these results suggest that the tyrosine residue at position 16 is necessary to constrain TCR reactivity for InsB9-23 by both limiting the development of pathogenic T cells and supporting the selection of Tregs.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Forkhead Transcription Factors/metabolism
- Insulin/genetics
- Insulin/immunology
- Mice
- Mice, Inbred NOD
- Mutation
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Maria Bettini
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- McNair Medical Institute, Texas Children's Hospital, Baylor College of Medicine, Houston, TX
| | - Marissa A Scavuzzo
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX
| | - Baoyu Liu
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Elizabeth Kolawole
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Lin Guo
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Brian D Evavold
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Malgorzata Borowiak
- McNair Medical Institute, Texas Children's Hospital, Baylor College of Medicine, Houston, TX
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX
- Department of Molecular and Cellular Biology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Matthew L Bettini
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- McNair Medical Institute, Texas Children's Hospital, Baylor College of Medicine, Houston, TX
| |
Collapse
|
120
|
Liu B, Hood JD, Kolawole EM, Woodruff DM, Vignali DA, Bettini M, Evavold BD. A Hybrid Insulin Epitope Maintains High 2D Affinity for Diabetogenic T Cells in the Periphery. Diabetes 2020; 69:381-391. [PMID: 31806623 PMCID: PMC7034185 DOI: 10.2337/db19-0399] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/26/2019] [Indexed: 12/20/2022]
Abstract
β-Cell antigen recognition by autoreactive T cells is essential in type 1 diabetes (T1D) pathogenesis. Recently, insulin hybrid peptides (HIPs) were identified as strong agonists for CD4 diabetogenic T cells. Here, using BDC2.5 transgenic and NOD mice, we investigated T-cell recognition of the HIP2.5 epitope, which is a fusion of insulin C-peptide and chromogranin A (ChgA) fragments, and compared it with the WE14 and ChgA29 -42 epitopes. We measured in situ two-dimensional affinity on individual live T cells from thymus, spleen, pancreatic lymph nodes, and islets before and after diabetes. Although preselection BDC2.5 thymocytes possess higher affinity than splenic BDC2.5 T cells for all three epitopes, peripheral splenic T cells maintained high affinity only to the HIP2.5 epitope. In polyclonal NOD mice, a high frequency (∼40%) of HIP2.5-specific islet T cells were identified at both prediabetic and diabetic stages comprising two distinct high- and low-affinity populations that differed in affinity by 100-fold. This high frequency of high- and low-affinity HIP2.5 T cells in the islets potentially represents a major risk factor in diabetes pathogenesis.
Collapse
Affiliation(s)
- Baoyu Liu
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Jennifer D Hood
- Department of Microbiology and Immunology, Emory University, Atlanta, GA
| | - Elizabeth M Kolawole
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | | | - Dario A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Maria Bettini
- Department of Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, TX
| | - Brian D Evavold
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| |
Collapse
|
121
|
Xu X, Li H, Xu C. Structural understanding of T cell receptor triggering. Cell Mol Immunol 2020; 17:193-202. [PMID: 32047259 PMCID: PMC7052162 DOI: 10.1038/s41423-020-0367-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/08/2020] [Indexed: 11/09/2022] Open
Abstract
The T cell receptor (TCR) is one of the most complicated receptors in mammalian cells, and its triggering mechanism remains mysterious. As an octamer complex, TCR comprises an antigen-binding subunit (TCRαβ) and three CD3 signaling subunits (CD3ζζ, CD3δε, and CD3γε). Engagement of TCRαβ with an antigen peptide presented on the MHC leads to tyrosine phosphorylation of the immunoreceptor tyrosine-based activation motif (ITAM) in CD3 cytoplasmic domains (CDs), thus translating extracellular binding kinetics to intracellular signaling events. Whether conformational change plays an important role in the transmembrane signal transduction of TCR is under debate. Attracted by the complexity and functional importance of TCR, many groups have been studying TCR structure and triggering for decades using diverse biochemical and biophysical tools. Here, we synthesize these structural studies and discuss the relevance of the conformational change model in TCR triggering.
Collapse
Affiliation(s)
- Xinyi Xu
- State Key Laboratory of Molecular Biology, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, 200031, Shanghai, China
| | - Hua Li
- State Key Laboratory of Molecular Biology, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, 200031, Shanghai, China
| | - Chenqi Xu
- State Key Laboratory of Molecular Biology, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, 200031, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, 201210, Shanghai, China.
| |
Collapse
|
122
|
Blumenthal D, Burkhardt JK. Multiple actin networks coordinate mechanotransduction at the immunological synapse. J Cell Biol 2020; 219:e201911058. [PMID: 31977034 PMCID: PMC7041673 DOI: 10.1083/jcb.201911058] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/31/2019] [Accepted: 01/02/2020] [Indexed: 12/26/2022] Open
Abstract
Activation of naive T cells by antigen-presenting cells (APCs) is an essential step in mounting an adaptive immune response. It is known that antigen recognition and T cell receptor (TCR) signaling depend on forces applied by the T cell actin cytoskeleton, but until recently, the underlying mechanisms have been poorly defined. Here, we review recent advances in the field, which show that specific actin-dependent structures contribute to the process in distinct ways. In essence, T cell priming involves a tug-of-war between the cytoskeletons of the T cell and the APC, where the actin cytoskeleton serves as a mechanical intermediate that integrates force-dependent signals. We consider each of the relevant actin-rich T cell structures separately and address how they work together at the topologically and temporally complex cell-cell interface. In addition, we address how this mechanobiology can be incorporated into canonical immunological models to improve how these models explain T cell sensitivity and antigenic specificity.
Collapse
Affiliation(s)
| | - Janis K. Burkhardt
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
123
|
Obeidy P, Ju LA, Oehlers SH, Zulkhernain NS, Lee Q, Galeano Niño JL, Kwan RY, Tikoo S, Cavanagh LL, Mrass P, Cook AJ, Jackson SP, Biro M, Roediger B, Sixt M, Weninger W. Partial loss of actin nucleator actin-related protein 2/3 activity triggers blebbing in primary T lymphocytes. Immunol Cell Biol 2019; 98:93-113. [PMID: 31698518 PMCID: PMC7028084 DOI: 10.1111/imcb.12304] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 12/11/2022]
Abstract
T lymphocytes utilize amoeboid migration to navigate effectively within complex microenvironments. The precise rearrangement of the actin cytoskeleton required for cellular forward propulsion is mediated by actin regulators, including the actin‐related protein 2/3 (Arp2/3) complex, a macromolecular machine that nucleates branched actin filaments at the leading edge. The consequences of modulating Arp2/3 activity on the biophysical properties of the actomyosin cortex and downstream T cell function are incompletely understood. We report that even a moderate decrease of Arp3 levels in T cells profoundly affects actin cortex integrity. Reduction in total F‐actin content leads to reduced cortical tension and disrupted lamellipodia formation. Instead, in Arp3‐knockdown cells, the motility mode is dominated by blebbing migration characterized by transient, balloon‐like protrusions at the leading edge. Although this migration mode seems to be compatible with interstitial migration in three‐dimensional environments, diminished locomotion kinetics and impaired cytotoxicity interfere with optimal T cell function. These findings define the importance of finely tuned, Arp2/3‐dependent mechanophysical membrane integrity in cytotoxic effector T lymphocyte activities.
Collapse
Affiliation(s)
- Peyman Obeidy
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Lining A Ju
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW, 2006, Australia.,Heart Research Institute and Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program, The Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia.,Discipline of Infectious Diseases & Immunology, Marie Bashir Institute, The University of Sydney, Sydney, NSW, 2006, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Nursafwana S Zulkhernain
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Quintin Lee
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Jorge L Galeano Niño
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, the ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Kensington, NSW, 2033, Australia
| | - Rain Yq Kwan
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Shweta Tikoo
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Lois L Cavanagh
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Paulus Mrass
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Adam Jl Cook
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Shaun P Jackson
- Heart Research Institute and Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.,Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Maté Biro
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, the ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Kensington, NSW, 2033, Australia
| | - Ben Roediger
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Michael Sixt
- Institute of Science and Technology, Klosterneuburg, 3400, Austria
| | - Wolfgang Weninger
- Immune Imaging Program, The Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia.,Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia.,Department of Dermatology, Medical University of Vienna, Vienna, 1090, Austria
| |
Collapse
|
124
|
Kaitao L, William R, Zhou Y, Cheng Z. Single-molecule investigations of T-cell activation. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019; 12:102-110. [PMID: 32296738 PMCID: PMC7158867 DOI: 10.1016/j.cobme.2019.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
T-cell activation is the central event governing its development, differentiation, and effector functions. T-cell activation is initiated by the direct physical interaction of the T cell antigen receptor (TCR) with cognate peptide presented by the major histocompatibility complex (pMHC) molecule expressed on the antigen presenting cell (APC) surface. Since the identification of TCR as the only receptor for antigen on T cells three decades ago, studies have elucidated the major molecular players and signaling events responding to TCR stimulation. However, the question of how the physical event of pMHC binding is converted across the membrane into chemical events to initiate signal transduction remains elusive. Here we review recent investigations of T-cell activation using single-molecule force and fluorescence techniques that shed new light on this key question.
Collapse
Affiliation(s)
- Li Kaitao
- Wallace H. Coulter Department of Biomedical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Rittase William
- Wallace H. Coulter Department of Biomedical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Yuan Zhou
- George W. Woodruff School of Mechanical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Zhu Cheng
- Wallace H. Coulter Department of Biomedical Engineering
- George W. Woodruff School of Mechanical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
125
|
Grossman Z. Immunological Paradigms, Mechanisms, and Models: Conceptual Understanding Is a Prerequisite to Effective Modeling. Front Immunol 2019; 10:2522. [PMID: 31749803 PMCID: PMC6848063 DOI: 10.3389/fimmu.2019.02522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022] Open
Abstract
Most mathematical models that describe the individual or collective actions of cells aim at creating faithful representations of limited sets of data in a self-consistent manner. Consistency with relevant physiological rules pertaining to the greater picture is rarely imposed. By themselves, such models have limited predictive or even explanatory value, contrary to standard claims. Here I try to show that a more critical examination of currently held paradigms is necessary and could potentially lead to models that pass the test of time. In considering the evolution of paradigms over the past decades I focus on the “smart surveillance” theory of how T cells can respond differentially, individually and collectively, to both self- and foreign antigens depending on various “contextual” parameters. The overall perspective is that physiological messages to cells are encoded not only in the biochemical connections of signaling molecules to the cellular machinery but also in the magnitude, kinetics, and in the time- and space-contingencies, of sets of stimuli. By rationalizing the feasibility of subthreshold interactions, the “dynamic tuning hypothesis,” a central component of the theory, set the ground for further theoretical and experimental explorations of dynamically regulated immune tolerance, homeostasis and diversity, and of the notion that lymphocytes participate in nonclassical physiological functions. Some of these efforts are reviewed. Another focus of this review is the concomitant regulation of immune activation and homeostasis through the operation of a feedback mechanism controlling the balance between renewal and differentiation of activated cells. Different perspectives on the nature and regulation of chronic immune activation in HIV infection have led to conflicting models of HIV pathogenesis—a major area of research for theoretical immunologists over almost three decades—and can have profound impact on ongoing HIV cure strategies. Altogether, this critical review is intended to constructively influence the outlook of prospective model builders and of interested immunologists on the state of the art and to encourage conceptual work.
Collapse
Affiliation(s)
- Zvi Grossman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
126
|
Spear TT, Evavold BD, Baker BM, Nishimura MI. Understanding TCR affinity, antigen specificity, and cross-reactivity to improve TCR gene-modified T cells for cancer immunotherapy. Cancer Immunol Immunother 2019; 68:1881-1889. [PMID: 31595324 PMCID: PMC11028285 DOI: 10.1007/s00262-019-02401-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/20/2019] [Indexed: 12/19/2022]
Abstract
Adoptive cell transfer (ACT) using T cell receptor (TCR) gene-modified T cells is an exciting and rapidly evolving field. Numerous preclinical and clinical studies have demonstrated various levels of feasibility, safety, and efficacy using TCR-engineered T cells to treat cancer and viral infections. Although evidence suggests their use can be effective, to what extent and how to improve these therapeutics are still matters of investigation. As TCR affinity has been generally accepted as the central role in defining T cell specificity and sensitivity, selection for and generation of high affinity TCRs has remained a fundamental approach to design more potent T cells. However, traditional methods for affinity-enhancement by random mutagenesis can induce undesirable cross-reactivity causing on- and off-target adverse events, generate exhausted effectors by overstimulation, and ignore other kinetic and cellular parameters that have been shown to impact antigen specificity. In this Focussed Research Review, we comment on the preclinical and clinical potential of TCR gene-modified T cells, summarize our contributions challenging the role TCR affinity plays in antigen recognition, and explore how structure-guided design can be used to manipulate antigen specificity and TCR cross-reactivity to improve the safety and efficacy of TCR gene-modified T cells used in ACT.
Collapse
Affiliation(s)
- Timothy T Spear
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, 2160 S. 1st Ave, Bldg 112, Room 308, Maywood, IL, 60153, USA.
| | - Brian D Evavold
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Brian M Baker
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46530, USA
| | - Michael I Nishimura
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, 2160 S. 1st Ave, Bldg 112, Room 308, Maywood, IL, 60153, USA
| |
Collapse
|
127
|
Courtney AH, Shvets AA, Lu W, Griffante G, Mollenauer M, Horkova V, Lo WL, Yu S, Stepanek O, Chakraborty AK, Weiss A. CD45 functions as a signaling gatekeeper in T cells. Sci Signal 2019; 12:12/604/eaaw8151. [PMID: 31641081 DOI: 10.1126/scisignal.aaw8151] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
T cells require the protein tyrosine phosphatase CD45 to detect and respond to antigen because it activates the Src family kinase Lck, which phosphorylates the T cell antigen receptor (TCR) complex. CD45 activates Lck by opposing the negative regulatory kinase Csk. Paradoxically, CD45 has also been implicated in suppressing TCR signaling by dephosphorylating the same signaling motifs within the TCR complex upon which Lck acts. We sought to reconcile these observations using chemical and genetic perturbations of the Csk/CD45 regulatory axis incorporated with computational analyses. Specifically, we titrated the activities of Csk and CD45 and assessed their influence on Lck activation, TCR-associated ζ-chain phosphorylation, and more downstream signaling events. Acute inhibition of Csk revealed that CD45 suppressed ζ-chain phosphorylation and was necessary for a regulatable pool of active Lck, thereby interconnecting the activating and suppressive roles of CD45 that tune antigen discrimination. CD45 suppressed signaling events that were antigen independent or induced by low-affinity antigen but not those initiated by high-affinity antigen. Together, our findings reveal that CD45 acts as a signaling "gatekeeper," enabling graded signaling outputs while filtering weak or spurious signaling events.
Collapse
Affiliation(s)
- Adam H Courtney
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA. .,Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexey A Shvets
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wen Lu
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gloria Griffante
- Division of Molecular Immunology, Department of Internal Medicine, University Hospital Erlangen and Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | - Veronika Horkova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - Wan-Lin Lo
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Steven Yu
- Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA
| | - Ondrej Stepanek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - Arup K Chakraborty
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arthur Weiss
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA. .,Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA
| |
Collapse
|
128
|
Abstract
The immune response is orchestrated by a variety of immune cells. The function of each cell is determined by the collective signals from various immunoreceptors, whose expression and activity depend on the developmental stages of the cell and its environmental context. Recent studies have highlighted the presence of mechanical force on several immunoreceptor-ligand pairs and the important role of force in regulating their interaction and function. In this Perspective, we use the T cell antigen receptor as an example with which to review the current understanding of the mechanosensing properties of immunoreceptors. We discuss the types of forces that immunoreceptors may encounter and the effects of force on ligand bonding, conformational change and the triggering of immunoreceptors, as well as the effects of force on the downstream signal transduction, cell-fate decisions and effector function of immune cells.
Collapse
|
129
|
TCR-pMHC bond conformation controls TCR ligand discrimination. Cell Mol Immunol 2019; 17:203-217. [PMID: 31530899 PMCID: PMC7052167 DOI: 10.1038/s41423-019-0273-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 08/07/2019] [Indexed: 11/15/2022] Open
Abstract
A major unanswered question is how a TCR discriminates between foreign and self-peptides presented on the APC surface. Here, we used in situ fluorescence resonance energy transfer (FRET) to measure the distances of single TCR–pMHC bonds and the conformations of individual TCR–CD3ζ receptors at the membranes of live primary T cells. We found that a TCR discriminates between closely related peptides by forming single TCR–pMHC bonds with different conformations, and the most potent pMHC forms the shortest bond. The bond conformation is an intrinsic property that is independent of the binding affinity and kinetics, TCR microcluster formation, and CD4 binding. The bond conformation dictates the degree of CD3ζ dissociation from the inner leaflet of the plasma membrane via a positive calcium signaling feedback loop to precisely control the accessibility of CD3ζ ITAMs for phosphorylation. Our data revealed the mechanism by which a TCR deciphers the structural differences among peptides via the TCR–pMHC bond conformation.
Collapse
|
130
|
DNA probes that store mechanical information reveal transient piconewton forces applied by T cells. Proc Natl Acad Sci U S A 2019; 116:16949-16954. [PMID: 31391300 DOI: 10.1073/pnas.1904034116] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The advent of molecular tension probes for real-time mapping of piconewton forces in living systems has had a major impact on mechanobiology. For example, DNA-based tension probes have revealed roles for mechanics in platelet, B cell, T cell, and fibroblast function. Nonetheless, imaging short-lived forces transmitted by low-abundance receptors remains a challenge. This is a particular problem for mechanoimmunology where ligand-receptor bindings are short lived, and a few antigens are sufficient for cell triggering. Herein, we present a mechanoselection strategy that uses locking oligonucleotides to preferentially and irreversibly bind DNA probes that are mechanically strained over probes at rest. Thus, infrequent and short-lived mechanical events are tagged. This strategy allows for integration and storage of mechanical information into a map of molecular tension history. Upon addition of unlocking oligonucleotides that drive toehold-mediated strand displacement, the probes reset to the real-time state, thereby erasing stored mechanical information. As a proof of concept, we applied this strategy to study OT-1 T cells, revealing that the T cell receptor (TCR) mechanically samples antigens carrying single amino acid mutations. Such events are not detectable using conventional tension probes. Each mutant peptide ligand displayed a different level of mechanical sampling and spatial scanning by the TCR that strongly correlated with its functional potency. Finally, we show evidence that T cells transmit pN forces through the programmed cell death receptor-1 (PD1), a major target in cancer immunotherapy. We anticipate that mechanical information storage will be broadly useful in studying the mechanobiology of the immune system.
Collapse
|
131
|
de Oliveira LR, Jaqaman K. FISIK: Framework for the Inference of In Situ Interaction Kinetics from Single-Molecule Imaging Data. Biophys J 2019; 117:1012-1028. [PMID: 31443908 DOI: 10.1016/j.bpj.2019.07.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/27/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Recent experimental and computational developments have been pushing the limits of live-cell single-molecule imaging, enabling the monitoring of intermolecular interactions in their native environment with high spatiotemporal resolution. However, interactions are captured only for the labeled subset of molecules, which tends to be a small fraction. As a result, it has remained a challenge to calculate molecular interaction kinetics, in particular association rates, from live-cell single-molecule tracking data. To overcome this challenge, we developed a mathematical modeling-based Framework for the Inference of in Situ Interaction Kinetics (FISIK) from single-molecule imaging data with substoichiometric labeling. FISIK consists of (I) devising a mathematical model of molecular movement and interactions, mimicking the biological system and data-acquisition setup, and (II) estimating the unknown model parameters, including molecular association and dissociation rates, by fitting the model to experimental single-molecule data. Due to the stochastic nature of the model and data, we adapted the method of indirect inference for model calibration. We validated FISIK using a series of tests in which we simulated trajectories of diffusing molecules that interact with each other, considering a wide range of model parameters, and including resolution limitations, tracking errors, and mismatches between the model and the biological system it mimics. We found that FISIK has the sensitivity to determine association and dissociation rates, with accuracy and precision depending on the labeled fraction of molecules and the extent of molecule tracking errors. For cases where the labeled fraction is too low (e.g., to afford accurate tracking), combining dynamic but sparse single-molecule imaging data with almost-whole population oligomer distribution data improves FISIK's performance. All in all, FISIK is a promising approach for the derivation of molecular interaction kinetics in their native environment from single-molecule imaging data with substoichiometric labeling.
Collapse
Affiliation(s)
| | - Khuloud Jaqaman
- Department of Biophysics, UT Southwestern Medical Center, Dallas, Texas; Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
132
|
|
133
|
Can single molecule localization microscopy detect nanoclusters in T cells? Curr Opin Chem Biol 2019; 51:130-137. [DOI: 10.1016/j.cbpa.2019.05.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 05/10/2019] [Accepted: 05/21/2019] [Indexed: 11/21/2022]
|
134
|
He L, Raddatz AD, Zhou F, Hwang H, Kemp ML, Lu H. Dynamic Mitochondrial Migratory Features Associated with Calcium Responses during T Cell Antigen Recognition. THE JOURNAL OF IMMUNOLOGY 2019; 203:760-768. [PMID: 31201236 DOI: 10.4049/jimmunol.1800299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/20/2019] [Indexed: 01/09/2023]
Abstract
A T cell clone is able to distinguish Ags in the form of peptide-MHC complexes with high specificity and sensitivity; however, how subtle differences in peptide-MHC structures translate to distinct T cell effector functions remains unknown. We hypothesized that mitochondrial positioning and associated calcium responses play an important role in T cell Ag recognition. We engineered a microfluidic system to precisely manipulate and synchronize a large number of cell-cell pairing events, which provided simultaneous real-time signaling imaging and organelle tracking with temporal precision. In addition, we developed image-derived metrics to quantify calcium response and mitochondria movement. Using myelin proteolipid altered peptide ligands and a hybridoma T cell line derived from a mouse model of experimental autoimmune encephalomyelitis, we observed that Ag potency modulates calcium response at the single-cell level. We further developed a partial least squares regression model, which highlighted mitochondrial positioning as a strong predictor of calcium response. The model revealed T cell mitochondria sharply alter direction within minutes following exposure to agonist peptide Ag, changing from accumulation at the immunological synapse to retrograde movement toward the distal end of the T cell body. By quantifying mitochondria movement as a highly dynamic process with rapidly changing phases, our result reconciles conflicting prior reports of mitochondria positioning during T cell Ag recognition. We envision applying this pipeline of methodology to study cell interactions between other immune cell types to reveal important signaling phenomenon that is inaccessible because of data-limited experimental design.
Collapse
Affiliation(s)
- Luye He
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Andrew D Raddatz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Fangyuan Zhou
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332; and
| | - Hyundoo Hwang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Melissa L Kemp
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332; .,Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Hang Lu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332; .,Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
135
|
Chow IT, Gates TJ, Papadopoulos GK, Moustakas AK, Kolawole EM, Notturno RJ, McGinty JW, Torres-Chinn N, James EA, Greenbaum C, Nepom GT, Evavold BD, Kwok WW. Discriminative T cell recognition of cross-reactive islet-antigens is associated with HLA-DQ8 transdimer-mediated autoimmune diabetes. SCIENCE ADVANCES 2019; 5:eaaw9336. [PMID: 31457096 PMCID: PMC6703875 DOI: 10.1126/sciadv.aaw9336] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/11/2019] [Indexed: 05/04/2023]
Abstract
Human leukocyte antigen (HLA)-DQ8 transdimer (HLA-DQA1*0501/DQB1*0302) confers exceptionally high risk in autoimmune diabetes. However, little is known about HLA-DQ8 transdimer-restricted CD4 T cell recognition, an event crucial for triggering HLA-DQ8 transdimer-specific anti-islet immunity. Here, we report a high degree of epitope overlap and T cell promiscuity between susceptible HLA-DQ8 and HLA-DQ8 transdimer. Despite preservation of putative residues for T cell receptor (TCR) contact, stronger disease-associated responses to cross-reactive, immunodominant islet epitopes are elicited by HLA-DQ8 transdimer. Mutagenesis at the α chain of HLA-DQ8 transdimer in complex with the disease-relevant GAD65250-266 peptide and in silico analysis reveal the DQ α52 residue located within the N-terminal edge of the peptide-binding cleft for the enhanced T cell reactivity, altering avidity and biophysical affinity between TCR and HLA-peptide complexes. Accordingly, a structurally promiscuous but nondegenerate TCR-HLA-peptide interface is pivotal for HLA-DQ8 transdimer-mediated autoimmune diabetes.
Collapse
Affiliation(s)
- I-Ting Chow
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
| | - Theresa J. Gates
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
| | - George K. Papadopoulos
- Laboratory of Biophysics, Biochemistry, Bioprocessing and Bioproducts, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, GR47100 Arta, Greece
| | - Antonis K. Moustakas
- Department of Food Technology, Ionian University, GR28100 Argostoli, Cephallonia, Greece
| | - Elizabeth M. Kolawole
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Richard J. Notturno
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
| | - John W. McGinty
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
| | - Nadia Torres-Chinn
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
| | - Eddie A. James
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
| | - Carla Greenbaum
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
| | - Gerald T. Nepom
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Brian D. Evavold
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - William W. Kwok
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA 98101, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Corresponding author.
| |
Collapse
|
136
|
Membrane Organization and Physical Regulation of Lymphocyte Antigen Receptors: A Biophysicist's Perspective. J Membr Biol 2019; 252:397-412. [PMID: 31352492 DOI: 10.1007/s00232-019-00085-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/18/2019] [Indexed: 12/19/2022]
Abstract
Receptors at the membrane of immune cells are the central players of innate and adaptative immunity, providing effective defence mechanisms against pathogens or cancer cells. Their function is intimately linked to their position at and within the membrane which provides accessibility, mobility as well as membrane proximal cytoskeleton anchoring, all of these elements playing important roles in the final function and links to cellular actions. Understanding how immune cells integrate the specific signals received at their membrane to take a decision remains an immense challenge and a very active field of fundamental and applied research. Recent progress in imaging and micromanipulation techniques have led to an unprecedented refinement in the description of molecular structures and supramolecular assemblies at the immune cell membrane, and provided a glimpse into their dynamics and regulation by force. Several key elements have been scrutinized such as the roles of relative sizes of molecules, lateral organisation, motion in the membrane of the receptors, but also physical cues such as forces, mediated by cellular substrates of different rigidities or applied by the cell itself, in conjunction with its partner cell. We review here these recent discoveries associated with a description of the biophysical methods used. While a conclusive picture integrating all of these components is still lacking, mainly due to the implication of diverse and different mechanisms and spatio-temporal scales involved, the amount of quantitative data available opens the way for physical modelling and numerical simulations and new avenues for experimental research.
Collapse
|
137
|
Hu W, Zhang Y, Sun X, Zhang T, Xu L, Xie H, Li Z, Liu W, Lou J, Chen W. FcγRIIB-I232T polymorphic change allosterically suppresses ligand binding. eLife 2019; 8:46689. [PMID: 31343409 PMCID: PMC6711707 DOI: 10.7554/elife.46689] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/25/2019] [Indexed: 01/02/2023] Open
Abstract
FcγRIIB binding to its ligand suppresses immune cell activation. A single-nucleotide polymorphic (SNP) change, I232T, in the transmembrane (TM) domain of FcγRIIB loses its suppressive function, which is clinically associated with systemic lupus erythematosus (SLE). Previously, we reported that I232T tilts FcγRIIB’s TM domain. In this study, combining with molecular dynamics simulations and single-cell FRET assay, we further reveal that such tilting by I232T unexpectedly bends the FcγRIIB’s ectodomain toward plasma membrane to allosterically impede FcγRIIB’s ligand association. I232T substitution reduces in situ two-dimensional binding affinities and association rates of FcγRIIB to interact with its ligands, IgG1, IgG2 and IgG3 by three to four folds. This allosteric regulation by an SNP provides an intrinsic molecular mechanism for the functional loss of FcγRIIB-I232T in SLE patients. Left unchecked the immune system can cause devastating damage to healthy tissue. To prevent this from happening, immune cells have built-in off switches that dampen their activation. One such switch is a protein called FcγRIIB that sits on the outer surface of immune cells and binds to proteins known as antibodies, which are produced as part of the immune response. Its role is to act as a brake on the immune system, and stop it from getting out of control. Overactive immune cells can lead to autoimmune diseases such as systemic lupus erythematosus, also known as SLE for short, which causes damage to the skin, joints and other organs. Previous work suggests that SLE is correlated with a specific mutation in the FcγRIIB gene, but it is unclear how the mutation and the disease are connected. Proteins are made out of building blocks called amino acids, which have different chemical properties. A swap of one amino acid for another can have big consequences for the structure of a protein. In the case of FcγRIIB, the mutation that correlates with SLE changes an amino acid called isoleucine for another called threonine. Isoleucine does not mix well with water and is commonly found buried in the middle of proteins or inside cell membranes. Threonine, on the other hand, can readily interact with the hydrogen atoms in water and other amino acids. Hu, Zhang, Sun et al. used computer simulations and imaged single human cells to find out how the isoleucine to threonine change causes immune cells to become over-activated. The experiments revealed that threonine interacts with a nearby amino acid, putting a kink in the FcγRIIB protein. This kink causes the outer part of the FcγRIIB protein to bend towards the immune cell membrane, stopping it from binding to antibodies, and putting a break on immune cells that have become hyper-activated. There is currently no cure for SLE, but understanding its causes could take us a step closer to better management of the disease. Small molecule drug treatments often target the three-dimensional shape of certain proteins, so understanding the effect of mutations at the molecular level could help with the design of new treatments in the future.
Collapse
Affiliation(s)
- Wei Hu
- Department of Neurobiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Zhang
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaolin Sun
- Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Department of Rheumatology and Immunology, Peking-Tsinghua Center for Life Sciences, Peking University People's Hospital, Beijing, China
| | - Tongtong Zhang
- Department of Neurobiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liling Xu
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, China
| | - Hengyi Xie
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, China
| | - Zhanguo Li
- Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Department of Rheumatology and Immunology, Peking-Tsinghua Center for Life Sciences, Peking University People's Hospital, Beijing, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, China
| | - Jizhong Lou
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Chen
- Department of Neurobiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory for Biomedical Engineering of Ministry of Education, State Key Laboratory for Modern Optical Instrumentation, College of Biomedical Engineering and Instrument Science, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| |
Collapse
|
138
|
TCR-pMHC kinetics under force in a cell-free system show no intrinsic catch bond, but a minimal encounter duration before binding. Proc Natl Acad Sci U S A 2019; 116:16943-16948. [PMID: 31315981 DOI: 10.1073/pnas.1902141116] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The T cell receptor (TCR)-peptide-MHC (pMHC) interaction is the only antigen-specific interaction during T lymphocyte activation. Recent work suggests that formation of catch bonds is characteristic of activating TCR-pMHC interactions. However, whether this binding behavior is an intrinsic feature of the molecular bond, or a consequence of more complex multimolecular or cellular responses, remains unclear. We used a laminar flow chamber to measure, first, 2D TCR-pMHC dissociation kinetics of peptides of various activating potency in a cell-free system in the force range (6 to 15 pN) previously associated with catch-slip transitions and, second, 2D TCR-pMHC association kinetics, for which the method is well suited. We did not observe catch bonds in dissociation, and the off-rate measured in the 6- to 15-pN range correlated well with activation potency, suggesting that formation of catch bonds is not an intrinsic feature of the TCR-pMHC interaction. The association kinetics were better explained by a model with a minimal encounter duration rather than a standard on-rate constant, suggesting that membrane fluidity and dynamics may strongly influence bond formation.
Collapse
|
139
|
Fernandes RA, Ganzinger KA, Tzou JC, Jönsson P, Lee SF, Palayret M, Santos AM, Carr AR, Ponjavic A, Chang VT, Macleod C, Lagerholm BC, Lindsay AE, Dushek O, Tilevik A, Davis SJ, Klenerman D. A cell topography-based mechanism for ligand discrimination by the T cell receptor. Proc Natl Acad Sci U S A 2019; 116:14002-14010. [PMID: 31221762 PMCID: PMC6628812 DOI: 10.1073/pnas.1817255116] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The T cell receptor (TCR) initiates the elimination of pathogens and tumors by T cells. To avoid damage to the host, the receptor must be capable of discriminating between wild-type and mutated self and nonself peptide ligands presented by host cells. Exactly how the TCR does this is unknown. In resting T cells, the TCR is largely unphosphorylated due to the dominance of phosphatases over the kinases expressed at the cell surface. However, when agonist peptides are presented to the TCR by major histocompatibility complex proteins expressed by antigen-presenting cells (APCs), very fast receptor triggering, i.e., TCR phosphorylation, occurs. Recent work suggests that this depends on the local exclusion of the phosphatases from regions of contact of the T cells with the APCs. Here, we developed and tested a quantitative treatment of receptor triggering reliant only on TCR dwell time in phosphatase-depleted cell contacts constrained in area by cell topography. Using the model and experimentally derived parameters, we found that ligand discrimination likely depends crucially on individual contacts being ∼200 nm in radius, matching the dimensions of the surface protrusions used by T cells to interrogate their targets. The model not only correctly predicted the relative signaling potencies of known agonists and nonagonists but also achieved this in the absence of kinetic proofreading. Our work provides a simple, quantitative, and predictive molecular framework for understanding why TCR triggering is so selective and fast and reveals that, for some receptors, cell topography likely influences signaling outcomes.
Collapse
Affiliation(s)
- Ricardo A Fernandes
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Kristina A Ganzinger
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Justin C Tzou
- Department of Applied & Computational Mathematics & Statistics, University of Notre Dame, Notre Dame, IN 46556
| | - Peter Jönsson
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Steven F Lee
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Matthieu Palayret
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Alexander R Carr
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Aleks Ponjavic
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Veronica T Chang
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Charlotte Macleod
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - B Christoffer Lagerholm
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Alan E Lindsay
- Mathematics Department, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE Oxford, United Kingdom
- Wolfson Centre for Mathematical Biology, University of Oxford, OX1 3RE Oxford, United Kingdom
| | - Andreas Tilevik
- School of Bioscience, University of Skövde, 541 28 Skövde, Sweden
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom;
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - David Klenerman
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom;
| |
Collapse
|
140
|
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
|
141
|
Pullen RH, Abel SM. Mechanical feedback enables catch bonds to selectively stabilize scanning microvilli at T-cell surfaces. Mol Biol Cell 2019; 30:2087-2095. [PMID: 31116687 PMCID: PMC6727777 DOI: 10.1091/mbc.e19-01-0048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
T-cells use microvilli to search the surfaces of antigen-presenting cells for antigenic ligands. The active motion of scanning microvilli provides a force-generating mechanism that is intriguing in light of single-molecule experiments showing that applied forces increase the lifetimes of stimulatory receptor–ligand bonds (catch-bond behavior). In this work, we introduce a theoretical framework to explore the motion of a microvillar tip above an antigen-presenting surface when receptors on the tip stochastically bind to ligands on the surface and dissociate from them in a force-dependent manner. Forces on receptor-ligand bonds impact the motion of the microvillus, leading to feedback between binding and microvillar motion. We use computer simulations to show that the average microvillar velocity varies in a ligand-dependent manner; that catch bonds generate responses in which some microvilli almost completely stop, while others move with a broad distribution of velocities; and that the frequency of stopping depends on the concentration of stimulatory ligands. Typically, a small number of catch bonds initially immobilize the microvillus, after which additional bonds accumulate and increase the cumulative receptor-engagement time. Our results demonstrate that catch bonds can selectively slow and stabilize scanning microvilli, suggesting a physical mechanism that may contribute to antigen discrimination by T-cells.
Collapse
Affiliation(s)
- Robert H Pullen
- Department of Chemical and Biomolecular Engineering, National Institute for Mathematical and Biological Synthesis, University of Tennessee, Knoxville, TN 37996
| | - Steven M Abel
- Department of Chemical and Biomolecular Engineering, National Institute for Mathematical and Biological Synthesis, University of Tennessee, Knoxville, TN 37996
| |
Collapse
|
142
|
Wan Z, Shaheen S, Chau A, Zeng Y, Liu W. Imaging: Gear up for mechano-immunology. Cell Immunol 2019; 350:103926. [PMID: 31151736 DOI: 10.1016/j.cellimm.2019.103926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 04/15/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022]
Abstract
Immune cells including B and T lymphocytes have a remarkable ability to sense the physical perturbations through their surface expressed receptors. At the advent of modern imaging technologies paired with biophysical methods, we have gained the understanding of mechanical forces exerted by immune cells to perform their functions. This review will go over the imaging techniques already being used to study mechanical forces in immune cells. We will also discuss the dire need for new modern technologies for future work.
Collapse
Affiliation(s)
- Zhengpeng Wan
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Samina Shaheen
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Alicia Chau
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Yingyue Zeng
- School of Life Science, Liaoning University, Shenyang 110036, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
143
|
Nelson N, Lopez-Pelaez M, Palazon A, Poon E, De La Roche M, Barry S, Valge-Archer V, Wilkinson RW, Dovedi SJ, Smith PD. A cell-engineered system to assess tumor cell sensitivity to CD8 + T cell-mediated cytotoxicity. Oncoimmunology 2019; 8:1599635. [PMID: 31413906 PMCID: PMC6682348 DOI: 10.1080/2162402x.2019.1599635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/08/2019] [Accepted: 03/18/2019] [Indexed: 01/23/2023] Open
Abstract
In vitro assays that evaluate CD8+ T cell-mediated cytotoxicity are important to aid in the development of novel therapeutic approaches to enhance anti-tumor immune responses. Here, we describe a novel cytotoxicity co-culture assay that circumvents the problem of highly variable allogeneic responses and obviates the constraints of HLA-restriction between effector and target cells. We show that this assay can be easily applied to a panel of tumor cell lines to provide additional insights into intrinsic drivers of sensitivity/resistance to T cell-mediated killing, and to evaluate the impact of targeted therapies on both tumor and T cell compartments.
Collapse
Affiliation(s)
- Nadine Nelson
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | | | | | - Maike De La Roche
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Simon Barry
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | | | | | - Paul D. Smith
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| |
Collapse
|
144
|
Li L, Hu J, Li L, Song F. Binding constant of membrane-anchored receptors and ligands that induce membrane curvatures. SOFT MATTER 2019; 15:3507-3514. [PMID: 30912540 DOI: 10.1039/c8sm02504e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cell adhesion is crucial for immune response, tissue formation, and cell locomotion. The adhesion process is mediated by the specific binding of membrane-anchored receptor and ligand proteins. These adhesion proteins are in contact with the membranes and may generate curvature, which has been shown for a number of membrane proteins to play an important role in membrane remodeling. An important question remains of whether the local membrane curvatures induced by the adhesion proteins affect their binding. We've performed Monte Carlo simulations of a mesoscopic model for membrane adhesion via the specific binding of curvature-inducing receptors and ligands. We find that the curvatures induced by the adhesion proteins do affect their binding equilibrium constant. We presented a theory that takes into account the membrane deformations and protein-protein interactions due to the induced curvatures, and agrees quantitatively with our simulation results. Our study suggests that the ability to induce membrane curvatures represents a molecular property of the adhesion proteins and should be carefully considered in experimental characterization of the binding affinity.
Collapse
Affiliation(s)
- Long Li
- State Key Laboratory of Nonlinear Mechanics (LNM) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.
| | | | | | | |
Collapse
|
145
|
Shwetank, Frost EL, Mockus TE, Ren HM, Toprak M, Lauver MD, Netherby-Winslow CS, Jin G, Cosby JM, Evavold BD, Lukacher AE. PD-1 Dynamically Regulates Inflammation and Development of Brain-Resident Memory CD8 T Cells During Persistent Viral Encephalitis. Front Immunol 2019; 10:783. [PMID: 31105690 PMCID: PMC6499176 DOI: 10.3389/fimmu.2019.00783] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/25/2019] [Indexed: 01/07/2023] Open
Abstract
Programmed cell death-1 (PD-1) receptor signaling dampens the functionality of T cells faced with repetitive antigenic stimulation from chronic infections or tumors. Using intracerebral (i.c.) inoculation with mouse polyomavirus (MuPyV), we have shown that CD8 T cells establish a PD-1hi, tissue-resident memory population in the brains (bTRM) of mice with a low-level persistent infection. In MuPyV encephalitis, PD-L1 was expressed on infiltrating myeloid cells, microglia and astrocytes, but not on oligodendrocytes. Engagement of PD-1 on anti-MuPyV CD8 T cells limited their effector activity. NanoString gene expression analysis showed that neuroinflammation was higher in PD-L1-/- than wild type mice at day 8 post-infection, the peak of the MuPyV-specific CD8 response. During the persistent phase of infection, however, the absence of PD-1 signaling was found to be associated with a lower inflammatory response than in wild type mice. Genetic disruption and intracerebroventricular blockade of PD-1 signaling resulted in an increase in number of MuPyV-specific CD8 bTRM and the fraction of these cells expressing CD103, the αE integrin commonly used to define tissue-resident T cells. However, PD-L1-/- mice persistently infected with MuPyV showed impaired virus control upon i.c. re-infection with MuPyV. Collectively, these data reveal a temporal duality in PD-1-mediated regulation of MuPyV-associated neuroinflammation. PD-1 signaling limited the severity of neuroinflammation during acute infection but sustained a level of inflammation during persistent infection for maintaining control of virus re-infection.
Collapse
Affiliation(s)
- Shwetank
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, United States
| | - Elizabeth L. Frost
- Immunology and Molecular Pathogenesis Graduate Program, Emory University, Atlanta, GA, United States
| | - Taryn E. Mockus
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, United States
| | - Heather M. Ren
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, United States
| | - Mesut Toprak
- Section of Neuropathology, Yale School of Medicine, New Haven, CT, United States
| | - Matthew D. Lauver
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, United States
| | | | - Ge Jin
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, United States
| | - Jennifer M. Cosby
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | - Brian D. Evavold
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, United States,*Correspondence: Aron E. Lukacher
| |
Collapse
|
146
|
Tischer DK, Weiner OD. Light-based tuning of ligand half-life supports kinetic proofreading model of T cell signaling. eLife 2019; 8:42498. [PMID: 30947808 PMCID: PMC6488292 DOI: 10.7554/elife.42498] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 04/03/2019] [Indexed: 11/30/2022] Open
Abstract
T cells are thought to discriminate self from foreign peptides by converting small differences in ligand binding half-life into large changes in cell signaling. Such a kinetic proofreading model has been difficult to test directly, as existing methods of altering ligand binding half-life also change other potentially important biophysical parameters, most notably the mechanical stability of the receptor-ligand interaction. Here we develop an optogenetic approach to specifically tune the binding half-life of a chimeric antigen receptor without changing other binding parameters and provide direct evidence of kinetic proofreading in T cell signaling. This half-life discrimination is executed in the proximal signaling pathway, downstream of ZAP70 recruitment and upstream of diacylglycerol accumulation. Our methods represent a general tool for temporal and spatial control of T cell signaling and extend the reach of optogenetics to probe pathways where the individual molecular kinetics, rather than the ensemble average, gates downstream signaling.
Collapse
Affiliation(s)
- Doug K Tischer
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Orion David Weiner
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
147
|
Harrison DL, Fang Y, Huang J. T-Cell Mechanobiology: Force Sensation, Potentiation, and Translation. FRONTIERS IN PHYSICS 2019; 7:45. [PMID: 32601597 PMCID: PMC7323161 DOI: 10.3389/fphy.2019.00045] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
A T cell is a sensitive self-referential mechanical sensor. Mechanical forces influence the recognition, activation, differentiation, and function throughout the lifetime of a T cell. T cells constantly perceive and respond to physical stimuli through their surface receptors, cytoskeleton, and subcellular structures. Surface receptors receive physical cues in the form of forces generated through receptor-ligand binding events, which are dynamically regulated by contact tension, shear stress, and substrate rigidity. The resulting mechanotransduction not only influences T-cell recognition and signaling but also possibly modulates cell metabolism and gene expression. Moreover, forces also dynamically regulate the deformation, organization, and translocation of cytoskeleton and subcellular structures, leading to changes in T-cell mobility, migration, and infiltration. However, the roles and mechanisms of how mechanical forces modulate T-cell recognition, signaling, metabolism, and gene expression, are largely unknown and underappreciated. Here, we review recent technological and scientific advances in T-cell mechanobiology, discuss possible roles and mechanisms of T-cell mechanotransduction, and propose new research directions of this emerging field in health and disease.
Collapse
Affiliation(s)
- Devin L. Harrison
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
| | - Yun Fang
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, United States
| | - Jun Huang
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
- Institute for Molecular Engineering, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
148
|
Gálvez J, Gálvez JJ, García-Peñarrubia P. Is TCR/pMHC Affinity a Good Estimate of the T-cell Response? An Answer Based on Predictions From 12 Phenotypic Models. Front Immunol 2019; 10:349. [PMID: 30886616 PMCID: PMC6410681 DOI: 10.3389/fimmu.2019.00349] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/11/2019] [Indexed: 11/13/2022] Open
Abstract
On the T-cell surface the TCR is the only molecule that senses antigen, and the engagement of TCR with its specific antigenic peptide (agonist)/MHC complex (pMHC) is determined by the biochemical parameters of the TCR-pMHC interaction. This interaction is the keystone of the adaptive immune response by triggering intracellular signaling pathways that induce the expression of genes required for T cell-mediated effector functions, such as T cell proliferation, cytokine secretion and cytotoxicity. To study the TCR-pMHC interaction one of its properties most extensively analyzed has been TCR-pMHC affinity. However, and despite of intensive experimental research, the results obtained are far from conclusive. Here, to determine if TCR-pMHC affinity is a reliable parameter to characterize T-cell responses, a systematic study has been performed based on the predictions of 12 phenotypic models. This approach has the advantage that allow us to study the response of a given system as a function of only those parameters in which we are interested while other system parameters remain constant. A little surprising, only the simple occupancy model predicts a direct relationship between affinity and response so that an increase in affinity always leads to larger responses. Conversely, in the others more elaborate models this clear situation does not occur, i.e., that a general positive correlation between affinity and immune response does not exist. This is mainly because affinity values are given by the quotient k on/k off where k on and k off are the rate constants of the binding process (i.e., affinity is in fact the quotient of two parameters), so that different sets of these rate constants can give the same value of affinity. However, except in the occupancy model, the predicted T-cell responses depend on the individual values of k on and k off rather than on their quotient k on/k off. This allows: a) that systems with the same affinity can show quite different responses; and b) that systems with low affinity may exhibit larger responses than systems with higher affinities. This would make affinity a poor estimate of T-cell responses and, as a result, data correlations between affinity and immune response should be interpreted and used with caution.
Collapse
Affiliation(s)
- Jesús Gálvez
- Department of Physical Chemistry, Faculty of Chemistry, University of Murcia, Murcia, Spain
| | - Juan J Gálvez
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Pilar García-Peñarrubia
- Department of Biochemistry and Molecular Biology B and Immunology, School of Medicine, University of Murcia, Murcia, Spain
| |
Collapse
|
149
|
Goyette J, Nieves DJ, Ma Y, Gaus K. How does T cell receptor clustering impact on signal transduction? J Cell Sci 2019; 132:132/4/jcs226423. [DOI: 10.1242/jcs.226423] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
ABSTRACT
The essential function of the T cell receptor (TCR) is to translate the engagement of peptides on the major histocompatibility complex (pMHC) into appropriate intracellular signals through the associated cluster of differentiation 3 (CD3) complex. The spatial organization of the TCR–CD3 complex in the membrane is thought to be a key regulatory element of signal transduction, raising the question of how receptor clustering impacts on TCR triggering. How signal transduction at the TCR–CD3 complex encodes the quality and quantity of pMHC molecules is not fully understood. This question can be approached by reconstituting T cell signaling in model and cell membranes and addressed by single-molecule imaging of endogenous proteins in T cells. We highlight such methods and further discuss how TCR clustering could affect pMHC rebinding rates, the local balance between kinase and phosphatase activity and/or the lipid environment to regulate the signal efficiency of the TCR–CD3 complex. We also examine whether clustering could affect the conformation of cytoplasmic CD3 tails through a biophysical mechanism. Taken together, we highlight how the spatial organization of the TCR–CD3 complex – addressed by reconstitution approaches – has emerged as a key regulatory element in signal transduction of this archetypal immune receptor.
Collapse
Affiliation(s)
- Jesse Goyette
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular imaging, University of New South Wales, Sydney 2052, Australia
| | - Daniel J. Nieves
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular imaging, University of New South Wales, Sydney 2052, Australia
| | - Yuanqing Ma
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular imaging, University of New South Wales, Sydney 2052, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular imaging, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
150
|
Jiang N, Schonnesen AA, Ma KY. Ushering in Integrated T Cell Repertoire Profiling in Cancer. Trends Cancer 2019; 5:85-94. [PMID: 30755308 PMCID: PMC6544389 DOI: 10.1016/j.trecan.2018.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/28/2022]
Abstract
Advances in immune profiling techniques have dramatically changed the cancer immunotherapy and monitoring landscape. High-throughput protein and gene expression technologies have paved the way for the discovery of therapeutic targets and biomarkers, and have made monitoring therapeutic response possible through the ability to independently assay the phenotype, specificity, exhaustion status, and lineage of single T cells. Although valuable insights into response profiling have been gained with current technologies, it has become evident that single-method profiling is insufficient to accurately capture an antitumor T cell response. We discuss and propose new methods that combine multiple axes of analysis to provide a comprehensive analysis of T cell repertoire in the fight against cancer.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA; LIVESTRONG Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA.
| | - Alexandra A Schonnesen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Ke-Yue Ma
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|