1
|
Rollins ZA, Curtis MB, George SC, Faller R. A Computational Strategy for the Rapid Identification and Ranking of Patient-Specific T Cell Receptors Bound to Neoantigens. Macromol Rapid Commun 2024; 45:e2400225. [PMID: 38839076 PMCID: PMC11661661 DOI: 10.1002/marc.202400225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/02/2024] [Indexed: 06/07/2024]
Abstract
T cell receptor (TCR) recognition of a peptide-major histocompatibility complex (pMHC) is crucial for adaptive immune response. The identification of therapeutically relevant TCR-pMHC protein pairs is a bottleneck in the implementation of TCR-based immunotherapies. The ability to computationally design TCRs to target a specific pMHC requires automated integration of next-generation sequencing, protein-protein structure prediction, molecular dynamics, and TCR ranking. A pipeline to evaluate patient-specific, sequence-based TCRs to a target pMHC is presented. Using the three most frequently expressed TCRs from 16 colorectal cancer patients, the protein-protein structure of the TCRs to the target CEA peptide-MHC is predicted using Modeller and ColabFold. TCR-pMHC structures are compared using automated equilibration and successive analysis. ColabFold generated configurations require an ≈2.5× reduction in equilibration time of TCR-pMHC structures compared to Modeller. The structural differences between Modeller and ColabFold are demonstrated by root mean square deviation (≈0.20 nm) between clusters of equilibrated configurations, which impact the number of hydrogen bonds and Lennard-Jones contacts between the TCR and pMHC. TCR ranking criteria that may prioritize TCRs for evaluation of in vitro immunogenicity are identified, and this ranking is validated by comparing to state-of-the-art machine learning-based methods trained to predict the probability of TCR-pMHC binding.
Collapse
Affiliation(s)
- Zachary A. Rollins
- Department of Chemical EngineeringUniversity of CaliforniaDavis, 1 Shields Ave, Bainer HallDavisCA95616USA
| | - Matthew B. Curtis
- Department of Biomedical EngineeringUniversity of CaliforniaDavis, 451 E. Health Sciences Dr., GBSF 2303DavisCA95616USA
| | - Steven C. George
- Department of Biomedical EngineeringUniversity of CaliforniaDavis, 451 E. Health Sciences Dr., GBSF 2303DavisCA95616USA
| | - Roland Faller
- Department of Chemical EngineeringUniversity of CaliforniaDavis, 1 Shields Ave, Bainer HallDavisCA95616USA
- Department of Chemical EngineeringTexas Tech UniversityLubbockTX79409USA
| |
Collapse
|
2
|
Mo G, Lu X, Wu S, Zhu W. Strategies and rules for tuning TCR-derived therapy. Expert Rev Mol Med 2023; 26:e4. [PMID: 38095091 PMCID: PMC11062142 DOI: 10.1017/erm.2023.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/17/2023] [Accepted: 12/05/2023] [Indexed: 04/04/2024]
Abstract
Manipulation of T cells has revolutionized cancer immunotherapy. Notably, the use of T cells carrying engineered T cell receptors (TCR-T) offers a favourable therapeutic pathway, particularly in the treatment of solid tumours. However, major challenges such as limited clinical response efficacy, off-target effects and tumour immunosuppressive microenvironment have hindered the clinical translation of this approach. In this review, we mainly want to guide TCR-T investigators on several major issues they face in the treatment of solid tumours after obtaining specific TCR sequences: (1) whether we have to undergo affinity maturation or not, and what parameter we should use as a criterion for being more effective. (2) What modifications can be added to counteract the tumour inhibitory microenvironment to make our specific T cells to be more effective and what is the safety profile of such modifications? (3) What are the new forms and possibilities for TCR-T cell therapy in the future?
Collapse
Affiliation(s)
- Guoheng Mo
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sha Wu
- Department of Immunology/Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Wei Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
García-Sánchez M, Castro M, Faro J. B cell receptors and free antibodies have different antigen-binding kinetics. Proc Natl Acad Sci U S A 2023; 120:e2220669120. [PMID: 37616223 PMCID: PMC10469035 DOI: 10.1073/pnas.2220669120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/21/2023] [Indexed: 08/26/2023] Open
Abstract
Since the pioneering works of Berg and Purcell, discriminating between diffusion followed by binding has played a central role in understanding cell signaling. B cell receptors (BCR) and antibodies (Ab) challenge that simplified view as binding to the antigen follows after a chain of diffusion and rotations, including whole molecule rotation and independent tilts and twists of their Fab arms due to their Y-shaped structure and flexibility. In this paper, we combine analytical calculations with Brownian simulations to derive the first-passage times due to these three rotations positioning the Fab paratopes at a proper distance and orientation required for antigen binding. Our results indicate that when measuring Ab-Ag effective kinetic binding rates, using experimental methods in which the analyte is in solution only gives values proportional to the intrinsic binding rates, [Formula: see text], and [Formula: see text], for values of [Formula: see text] up to [Formula: see text]. Beyond that, a plateau of the effective 3D on rate between [Formula: see text] and [Formula: see text] is attained. Additionally, for BCR-Ag interactions, the effective 2D on and off binding rates can only be inferred from the corresponding effective 3D on and off rates for values of effective 3D on rates lower than [Formula: see text]. This is highly relevant when trying to relate BCR-antigen-binding strength and B cell response, especially during germinal center reactions. Therefore, there is a pressing need to reexamine our current understanding of the BCR-antigen kinetic rates in germinal centers using the latest experimental assays for BCR-Ag interactions.
Collapse
Affiliation(s)
- Miguel García-Sánchez
- Instituto de Investigación Tecnológica and Grupo Interdisciplinar de Sistemas Complejos, Universidad Pontificia Comillas, MadridE28015, Spain
| | - Mario Castro
- Instituto de Investigación Tecnológica and Grupo Interdisciplinar de Sistemas Complejos, Universidad Pontificia Comillas, MadridE28015, Spain
| | - José Faro
- Area of Immunology, Faculty of Biology, and Centro de Investigaciones Biomédicas (Biomedical Research Center), Universidade de Vigo, Vigo36310, Spain
- Instituto Gulbenkian de Ciência, Oeiras2781-901, Portugal
| |
Collapse
|
4
|
Moldovan L, Song CH, Chen YC, Wang HJ, Ju LA. Biomembrane force probe (BFP): Design, advancements, and recent applications to live-cell mechanobiology. EXPLORATION (BEIJING, CHINA) 2023; 3:20230004. [PMID: 37933233 PMCID: PMC10624387 DOI: 10.1002/exp.20230004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/18/2023] [Indexed: 11/08/2023]
Abstract
Mechanical forces play a vital role in biological processes at molecular and cellular levels, significantly impacting various diseases such as cancer, cardiovascular disease, and COVID-19. Recent advancements in dynamic force spectroscopy (DFS) techniques have enabled the application and measurement of forces and displacements with high resolutions, providing crucial insights into the mechanical pathways underlying these diseases. Among DFS techniques, the biomembrane force probe (BFP) stands out for its ability to measure bond kinetics and cellular mechanosensing with pico-newton and nano-meter resolutions. Here, a comprehensive overview of the classical BFP-DFS setup is presented and key advancements are emphasized, including the development of dual biomembrane force probe (dBFP) and fluorescence biomembrane force probe (fBFP). BFP-DFS allows us to investigate dynamic bond behaviors on living cells and significantly enhances the understanding of specific ligand-receptor axes mediated cell mechanosensing. The contributions of BFP-DFS to the fields of cancer biology, thrombosis, and inflammation are delved into, exploring its potential to elucidate novel therapeutic discoveries. Furthermore, future BFP upgrades aimed at improving output and feasibility are anticipated, emphasizing its growing importance in the field of cell mechanobiology. Although BFP-DFS remains a niche research modality, its impact on the expanding field of cell mechanobiology is immense.
Collapse
Affiliation(s)
- Laura Moldovan
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
- Charles Perkins CentreThe University of SydneyCamperdownNew South WalesAustralia
- Heart Research InstituteNewtownNew South WalesAustralia
| | - Caroline Haoran Song
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
- Charles Perkins CentreThe University of SydneyCamperdownNew South WalesAustralia
- Heart Research InstituteNewtownNew South WalesAustralia
- Sydney Nano Institute (Sydney Nano)The University of SydneyCamperdownNew South WalesAustralia
| | - Yiyao Catherine Chen
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
| | - Haoqing Jerry Wang
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
- Heart Research InstituteNewtownNew South WalesAustralia
- Sydney Nano Institute (Sydney Nano)The University of SydneyCamperdownNew South WalesAustralia
| | - Lining Arnold Ju
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
- Charles Perkins CentreThe University of SydneyCamperdownNew South WalesAustralia
- Heart Research InstituteNewtownNew South WalesAustralia
- Sydney Nano Institute (Sydney Nano)The University of SydneyCamperdownNew South WalesAustralia
| |
Collapse
|
5
|
Du R, Li L, Ji J, Fan Y. Receptor-Ligand Binding: Effect of Mechanical Factors. Int J Mol Sci 2023; 24:ijms24109062. [PMID: 37240408 DOI: 10.3390/ijms24109062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/20/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Gaining insight into the in situ receptor-ligand binding is pivotal for revealing the molecular mechanisms underlying the physiological and pathological processes and will contribute to drug discovery and biomedical application. An important issue involved is how the receptor-ligand binding responds to mechanical stimuli. This review aims to provide an overview of the current understanding of the effect of several representative mechanical factors, such as tension, shear stress, stretch, compression, and substrate stiffness on receptor-ligand binding, wherein the biomedical implications are focused. In addition, we highlight the importance of synergistic development of experimental and computational methods for fully understanding the in situ receptor-ligand binding, and further studies should focus on the coupling effects of these mechanical factors.
Collapse
Affiliation(s)
- Ruotian Du
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Jing Ji
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
6
|
Ide H, Aoshi T, Saito M, Espulgar WV, Briones JC, Hosokawa M, Matsunaga H, Arikawa K, Takeyama H, Koyama S, Takamatsu H, Tamiya E. Linking antigen specific T-cell dynamics in a microfluidic chip to single cell transcription patterns. Biochem Biophys Res Commun 2023; 657:8-15. [PMID: 36963175 DOI: 10.1016/j.bbrc.2023.03.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
A new non-invasive screening profile has been realized that can aid in determining T-cell activation state at single-cell level. Production of activated T-cells with good specificity and stable proliferation is greatly beneficial for advancing adoptive immunotherapy as innate immunological cells are not effective in recognizing and eliminating cancer as expected. The screening method is realized by relating intracellular Ca2+ intensity and motility of T-cells interacting with APC (Antigen Presenting Cells) in a microfluidic chip. The system is tested using APC pulsed with OVA257-264 peptide and its modified affinities (N4, Q4, T4 and V4), and the T-cells from OT-1 mice. In addition, single cell RNA sequencing reveals the activation states of the cells and the clusters from the derived profiles can be indicative of the T-cell activation state. The presented system here can be versatile for a comprehensive application to proceed with T-cell-based immunotherapy and screen the antigen-specific T-cells with excellent efficiency and high proliferation.
Collapse
Affiliation(s)
- Hiroki Ide
- Graduate School of Engineering Osaka Univ, Japan; PhotoBIO Lab, AIST-Osaka Univ, Japan
| | - Taiki Aoshi
- Research Institute for Microbial Diseases, Osaka Univ, Japan
| | - Masato Saito
- PhotoBIO Lab, AIST-Osaka Univ, Japan; Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
| | | | - Jonathan Campos Briones
- Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Masahito Hosokawa
- Department of Life Science and Medical Bioscience, Waseda Univ, Japan; CBBD-OIL, AIST-Waseda Univ, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda Univ, Japan; Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Koji Arikawa
- Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda Univ, Japan; CBBD-OIL, AIST-Waseda Univ, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda Univ, Japan; Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | | | | | - Eiichi Tamiya
- PhotoBIO Lab, AIST-Osaka Univ, Japan; Institute of Scientific and Industrial Research, Osaka University, Japan
| |
Collapse
|
7
|
Dendritic cell-mimicking scaffolds for ex vivo T cell expansion. Bioact Mater 2023; 21:241-252. [PMID: 36157246 PMCID: PMC9474324 DOI: 10.1016/j.bioactmat.2022.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/10/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022] Open
Abstract
We propose an ex vivo T cell expansion system that mimics natural antigen-presenting cells (APCs) for adoptive cell therapy (ACT). Microfiber scaffolds coated with dendritic cell (DC) membrane replicate physicochemical properties of dendritic cells specific for T cell activation such as rapid recognition by T cells, long duration of T cell tethering, and DC-specific co-stimulatory cues. The DC membrane-coated scaffold is first surface-immobilized with T cell stimulatory ligands, anti-CD3 (αCD3) and anti-CD28 (αCD28) antibodies, followed by adsorption of releasable interleukin-2 (IL-2). The scaffolds present both surface and soluble cues to T cells ex vivo in the same way that these cues are presented by natural APCs in vivo. We demonstrate that the DC-mimicking scaffold promotes greater polyclonal expansion of primary human T cells as compared to αCD3/αCD28-functionalized Dynabead. More importantly, major histocompatibility complex molecules derived from the DC membrane of the scaffold allow antigen-specific T cell expansion with target cell-specific killing ability. In addition, most of the expanded T cells (∼97%) can be harvested from the scaffold by density gradient centrifugation. Overall, the DC-mimicking scaffold offers a scalable, modular, and customizable platform for rapid expansion of highly functional T cells for ACT. The scaffold mimics physicochemical properties of natural antigen-presenting cells. The scaffold presents T cell stimulatory cues as antigen-presenting cell does. This platform supports both polyclonal and antigen-specific T cell expansion. This platform offers a large-scale manufacturing system for adoptive cell therapy.
Collapse
|
8
|
Zheng S, Zou M, Shao Y, Wu H, Wu H, Wang X. Two-dimensional measurements of receptor-ligand interactions. Front Mol Biosci 2023; 10:1154074. [PMID: 36876050 PMCID: PMC9981951 DOI: 10.3389/fmolb.2023.1154074] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
Gaining insight into the two-dimensional receptor-ligand interactions, which play a significant role in various pivotal biological processes such as immune response and cancer metastasis, will deepen our understanding of numerous physiological and pathological mechanisms and contribute to biomedical applications and drug design. A central issue involved is how to measure the in situ receptor-ligand binding kinetics. Here, we review several representative mechanical-based and fluorescence-based methods, and briefly discuss the strengths and weaknesses for each method. In addition, we emphasize the great importance of the combination of experimental and computational methods in studying the receptor-ligand interactions, and further studies should focus on the synergistic development of experimental and computational methods.
Collapse
Affiliation(s)
- Songjie Zheng
- Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Min Zou
- Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yingfeng Shao
- Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Huaping Wu
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Helong Wu
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Xiaohuan Wang
- Department of Rehabilitation Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
9
|
Rollins ZA, Huang J, Tagkopoulos I, Faller R, George SC. A Computational Algorithm to Assess the Physiochemical Determinants of T Cell Receptor Dissociation Kinetics. Comput Struct Biotechnol J 2022; 20:3473-3481. [PMID: 35860406 PMCID: PMC9278023 DOI: 10.1016/j.csbj.2022.06.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022] Open
Abstract
The rational design of T Cell Receptors (TCRs) for immunotherapy has stagnated due to a limited understanding of the dynamic physiochemical features of the TCR that elicit an immunogenic response. The physiochemical features of the TCR-peptide major histocompatibility complex (pMHC) bond dictate bond lifetime which, in turn, correlates with immunogenicity. Here, we: i) characterize the force-dependent dissociation kinetics of the bond between a TCR and a set of pMHC ligands using Steered Molecular Dynamics (SMD); and ii) implement a machine learning algorithm to identify which physiochemical features of the TCR govern dissociation kinetics. Our results demonstrate that the total number of hydrogen bonds between the CDR2β-MHC⍺(β), CDR1α-Peptide, and CDR3β-Peptide are critical features that determine bond lifetime.
Collapse
Affiliation(s)
| | - Jun Huang
- University of California, Davis, Davis, California, Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | | | | | - Steven C. George
- Department of Biomedical Engineering
- Corresponding author at: Department of Biomedical Engineering, 451 E. Health Sciences Drive, room 2315, University of California, Davis, Davis, CA 95616.
| |
Collapse
|
10
|
Watson VE, Faniel ML, Kamili NA, Krueger LD, Zhu C. Immune-mediated alopecias and their mechanobiological aspects. Cells Dev 2022; 170:203793. [PMID: 35649504 PMCID: PMC10681075 DOI: 10.1016/j.cdev.2022.203793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023]
Abstract
Alopecia is a non-specific term for hair loss clinically diagnosed by the hair loss pattern and histological analysis of patient scalp biopsies. The immune-mediated alopecia subtypes, including alopecia areata, lichen planopilaris, frontal fibrosing alopecia, and central centrifugal cicatricial alopecia, are common, significant forms of alopecia subtypes. For example, alopecia areata is the most common autoimmune disease with a lifetime incidence of approximately 2% of the world's population. In this perspective, we discuss major results from studies of immune-mediated alopecia subtypes. These studies suggest the key event in disease onset as the collapse in immune privilege, which alters the hair follicle microenvironment, e.g., upregulation of major histocompatibility complex molecules and increase of cytokine production, and results in immune cell infiltration, inflammatory responses, and damage of hair follicles. We note that previous studies have established that the hair follicle has a complex mechanical microenvironment, which may regulate the function of not only tissue cells but also immune cell infiltrates. This suggests a potential for mechanobiology to contribute to alopecia research by adding new methods, new approaches, and new ways of thinking, which is missing in the existing literature. To fill this a gap in the alopecia research space, we develop a mechanobiological hypothesis that alterations in the hair follicle microenvironment, specifically in the mechanically responsive tissues and cells, partially due to loss of immune privilege, may be contributors to disease pathology. We further focus our discussion on the potential for applying mechanoimmunology to the study of T cell infiltrates in the hair follicle, as they are considered primary contributors to alopecia pathology. To establish the connection between the mechanoimmunological hypothesis and immune-mediated alopecia subtypes, we discuss what is known about the role of T cells in immune-mediated alopecia subtypes, using the most extensively studied AA as our model.
Collapse
Affiliation(s)
- Valencia E Watson
- Wallace H. Coulter Department of Biomedical Engineering, USA; Bioengineering PhD Program, USA; Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Makala L Faniel
- Wallace H. Coulter Department of Biomedical Engineering, USA; Bioengineering PhD Program, USA; Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Loren D Krueger
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, USA; Bioengineering PhD Program, USA; Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
11
|
Rollins ZA, Faller R, George SC. Using Molecular Dynamics Simulations to Interrogate T Cell Receptor Non-Equilibrium Kinetics. Comput Struct Biotechnol J 2022; 20:2124-2133. [PMID: 35832631 PMCID: PMC9092387 DOI: 10.1016/j.csbj.2022.04.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022] Open
Abstract
Insights into the atomic-scale interaction of the T Cell Receptor with the peptide Major Histocompatibility Complex. Investigation of the physiochemical features that correspond with T Cell Receptor recognition during dynamic dissociation. Implications of force-dependent non-equilibrium kinetics on T Cell Receptor mechanosensing.
An atomic-scale mechanism of T Cell Receptor (TCR) mechanosensing of peptides in the binding groove of the peptide-major histocompatibility complex (pMHC) may inform the design of novel TCRs for immunotherapies. Using steered molecular dynamics simulations, our study demonstrates that mutations to peptides in the binding groove of the pMHC – which are known to discretely alter the T cell response to an antigen – alter the MHC conformation at equilibrium. This subsequently impacts the overall strength (duration and length) of the TCR-pMHC bond under constant load. Moreover, physiochemical features of the TCR-pMHC dynamic bond strength, such as hydrogen bonds and Lennard-Jones contacts, correlate with the immunogenic response elicited by the specific peptide in the MHC groove. Thus, formation of transient TCR-pMHC bonds is characteristic of immunogenic peptides, and steered molecular dynamics simulations can be used in the overall design strategy of TCRs for immunotherapies.
Collapse
Affiliation(s)
- Zachary A. Rollins
- Department of Chemical Engineering, University of California, Davis, 1 Shields Ave, Bainer Hall, Davis, CA 95616, United States
| | - Roland Faller
- Department of Chemical Engineering, University of California, Davis, 1 Shields Ave, Bainer Hall, Davis, CA 95616, United States
| | - Steven C. George
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Dr., GBSF 2303, Davis, CA 95616, United States
- Corresponding author at: Department of Biomedical Engineering, 451 E. Health Sciences Drive, Room 2315, University of California, Davis, CA 95616, United States.
| |
Collapse
|
12
|
Ma VPY, Hu Y, Kellner AV, Brockman JM, Velusamy A, Blanchard AT, Evavold BD, Alon R, Salaita K. The magnitude of LFA-1/ICAM-1 forces fine-tune TCR-triggered T cell activation. SCIENCE ADVANCES 2022; 8:eabg4485. [PMID: 35213231 PMCID: PMC8880789 DOI: 10.1126/sciadv.abg4485] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 12/15/2021] [Indexed: 05/15/2023]
Abstract
T cells defend against cancer and viral infections by rapidly scanning the surface of target cells seeking specific peptide antigens. This key process in adaptive immunity is sparked upon T cell receptor (TCR) binding of antigens within cell-cell junctions stabilized by integrin (LFA-1)/intercellular adhesion molecule-1 (ICAM-1) complexes. A long-standing question in this area is whether the forces transmitted through the LFA-1/ICAM-1 complex tune T cell signaling. Here, we use spectrally encoded DNA tension probes to reveal the first maps of LFA-1 and TCR forces generated by the T cell cytoskeleton upon antigen recognition. DNA probes that control the magnitude of LFA-1 force show that F>12 pN potentiates antigen-dependent T cell activation by enhancing T cell-substrate engagement. LFA-1/ICAM-1 mechanical events with F>12 pN also enhance the discriminatory power of the TCR when presented with near cognate antigens. Overall, our results show that T cells integrate multiple channels of mechanical information through different ligand-receptor pairs to tune function.
Collapse
Affiliation(s)
| | - Yuesong Hu
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Anna V. Kellner
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA 30332, USA
| | - Joshua M. Brockman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA 30332, USA
| | - Arventh Velusamy
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Aaron T. Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA 30332, USA
| | - Brian D. Evavold
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Ronen Alon
- Department of Immunology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
Dam T, Chouliara M, Junghans V, Jönsson P. Supported Lipid Bilayers and the Study of Two-Dimensional Binding Kinetics. Front Mol Biosci 2022; 9:833123. [PMID: 35252352 PMCID: PMC8896763 DOI: 10.3389/fmolb.2022.833123] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Binding between protein molecules on contacting cells is essential in initiating and regulating several key biological processes. In contrast to interactions between molecules in solution, these events are restricted to the two-dimensional (2D) plane of the meeting cell surfaces. However, converting between the more commonly available binding kinetics measured in solution and the so-called 2D binding kinetics has proven a complicated task since for the latter several factors other than the protein-protein interaction per se have an impact. A few important examples of these are: protein density, membrane fluctuations, force on the bond and the use of auxiliary binding molecules. The development of model membranes, and in particular supported lipid bilayers (SLBs), has made it possible to simplify the studied contact to analyze these effects and to measure 2D binding kinetics of individual protein-protein interactions. We will in this review give an overview of, and discuss, how different SLB systems have been used for this and compare different methods to measure binding kinetics in cell-SLB contacts. Typically, the SLB is functionalized with fluorescently labelled ligands whose interaction with the corresponding receptor on a binding cell can be detected. This interaction can either be studied 1) by an accumulation of ligands in the cell-SLB contact, whose magnitude depends on the density of the proteins and binding affinity of the interaction, or 2) by tracking single ligands in the SLB, which upon interaction with a receptor result in a change of motion of the diffusing ligand. The advantages and disadvantages of other methods measuring 2D binding kinetics will also be discussed and compared to the fluorescence-based methods. Although binding kinetic measurements in cell-SLB contacts have provided novel information on how ligands interact with receptors in vivo the number of these measurements is still limited. This is influenced by the complexity of the system as well as the required experimental time. Moreover, the outcome can vary significantly between studies, highlighting the necessity for continued development of methods to study 2D binding kinetics with higher precision and ease.
Collapse
Affiliation(s)
- Tommy Dam
- Department of Chemistry, Lund University, Lund, Sweden
| | | | - Victoria Junghans
- Nuffield Department of Medicine, CAMS Oxford Institute, University of Oxford, Oxford, United Kingdom
| | - Peter Jönsson
- Department of Chemistry, Lund University, Lund, Sweden
- *Correspondence: Peter Jönsson,
| |
Collapse
|
14
|
Motz CT, Kabat V, Saxena T, Bellamkonda RV, Zhu C. Neuromechanobiology: An Expanding Field Driven by the Force of Greater Focus. Adv Healthc Mater 2021; 10:e2100102. [PMID: 34342167 PMCID: PMC8497434 DOI: 10.1002/adhm.202100102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 07/06/2021] [Indexed: 12/14/2022]
Abstract
The brain processes information by transmitting signals through highly connected and dynamic networks of neurons. Neurons use specific cellular structures, including axons, dendrites and synapses, and specific molecules, including cell adhesion molecules, ion channels and chemical receptors to form, maintain and communicate among cells in the networks. These cellular and molecular processes take place in environments rich of mechanical cues, thus offering ample opportunities for mechanical regulation of neural development and function. Recent studies have suggested the importance of mechanical cues and their potential regulatory roles in the development and maintenance of these neuronal structures. Also suggested are the importance of mechanical cues and their potential regulatory roles in the interaction and function of molecules mediating the interneuronal communications. In this review, the current understanding is integrated and promising future directions of neuromechanobiology are suggested at the cellular and molecular levels. Several neuronal processes where mechanics likely plays a role are examined and how forces affect ligand binding, conformational change, and signal induction of molecules key to these neuronal processes are indicated, especially at the synapse. The disease relevance of neuromechanobiology as well as therapies and engineering solutions to neurological disorders stemmed from this emergent field of study are also discussed.
Collapse
Affiliation(s)
- Cara T Motz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Victoria Kabat
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Tarun Saxena
- Department of Biomedical Engineering, Duke University, Durham, NC, 27709, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| |
Collapse
|
15
|
Distinct roles of ICOS and CD40L in human T-B cell adhesion and antibody production. Cell Immunol 2021; 368:104420. [PMID: 34418679 DOI: 10.1016/j.cellimm.2021.104420] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/22/2021] [Accepted: 07/31/2021] [Indexed: 12/13/2022]
Abstract
CD40-CD40L and inducible co-stimulatory molecule (ICOS)-ICOSL ligations are demonstrated to play critical roles in CD4+T-B interaction for B cell activation and differentiation in mouse models. Herein, by using a micropipette adhesion assay and an in vitro CD4+T-B cell coculture system simultaneously, we intended to dissect their roles in human CD4+T-B adhesion and IgG/IgM production. With the upregulation of CD40L and ICOS expressions on CD4+ T cells upon TCR/CD28 stimulation in vitro, activated CD4+ T cells exhibited enhanced adhesion with autologous B cells at a single cell level when compared to the resting counterparts. Blockade of ICOS dramatically damped the adhesion between CD4+ T and B cells whereas less effect of CD40L blockade was observed. On the contrary, blockade of CD40L led to the dramatic decrease in IgG/IgM production when B cells were cocultured with activated CD4+ T cells together with the decrease in the induction of CD19hi B cells. However, ICOS blockade displayed less attenuation on IgG/IgM production. Distinct roles of CD40-CD40L and ICOS-ICOSL in cell adhesion and IgG/IgM production were also observed in CD4+T-B cell interaction in system lupus erythematosus patients. The blockade of CD40L, rather than ICOS, led to the dramatic decrease in the phosphorylation of Pyk2 in CD19hi B cells and total B cells. Our study thus provides the evidence that CD40L and ICOS on activated CD4+ T cells either upon in vitro activation or at the pathogenic state function diversely during CD4+T-B cell interactions. While ICOS-ICOSL ligation is more likely to be engaged in cell adhesion, CD40-CD40L provides indispensable signal for B cell differentiation and IgG/IgM production. Our results are thus indicative for the segregating costimulation of CD40-CD40L and ICOS-ICOSL on CD4+ T cells for B cell activation and differentiation, which might be helpful for the dissection of SLE pathogenesis.
Collapse
|
16
|
PD-1 suppresses TCR-CD8 cooperativity during T-cell antigen recognition. Nat Commun 2021; 12:2746. [PMID: 33980853 PMCID: PMC8115078 DOI: 10.1038/s41467-021-22965-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/09/2021] [Indexed: 12/31/2022] Open
Abstract
Despite the clinical success of blocking its interactions, how PD-1 inhibits T-cell activation is incompletely understood, as exemplified by its potency far exceeding what might be predicted from its affinity for PD-1 ligand-1 (PD-L1). This may be partially attributed to PD-1's targeting the proximal signaling of the T-cell receptor (TCR) and co-stimulatory receptor CD28 via activating Src homology region 2 domain-containing phosphatases (SHPs). Here, we report PD-1 signaling regulates the initial TCR antigen recognition manifested in a smaller spreading area, fewer molecular bonds formed, and shorter bond lifetime of T cell interaction with peptide-major histocompatibility complex (pMHC) in the presence than absence of PD-L1 in a manner dependent on SHPs and Leukocyte C-terminal Src kinase. Our results identify a PD-1 inhibitory mechanism that disrupts the cooperative TCR-pMHC-CD8 trimolecular interaction, which prevents CD8 from augmenting antigen recognition, explaining PD-1's potent inhibitory function and its value as a target for clinical intervention.
Collapse
|
17
|
Bingöl EN, Serçinoğlu O, Ozbek P. Unraveling the Allosteric Communication Mechanisms in T-Cell Receptor-Peptide-Loaded Major Histocompatibility Complex Dynamics Using Molecular Dynamics Simulations: An Approach Based on Dynamic Cross Correlation Maps and Residue Interaction Energy Calculations. J Chem Inf Model 2021; 61:2444-2453. [PMID: 33930270 DOI: 10.1021/acs.jcim.1c00338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antigen presentation by major histocompatibility complex (MHC) proteins to T-cell receptors (TCRs) plays a crucial role in triggering the adaptive immune response. Most of our knowledge on TCR-peptide-loaded major histocompatibility complex (pMHC) interaction stemmed from experiments yielding static structures, yet the dynamic aspects of this molecular interaction are equally important to understand the underlying molecular mechanisms and to develop treatment strategies against diseases such as cancer and autoimmune diseases. To this end, computational biophysics studies including all-atom molecular dynamics simulations have provided useful insights; however, we still lack a basic understanding of an overall allosteric mechanism that results in conformational changes in the TCR and subsequent T-cell activation. Previous hydrogen-deuterium exchange and nuclear magnetic resonance studies provided clues regarding these molecular mechanisms, including global rigidification and allosteric effects on the constant domain of TCRs away from the pMHC interaction site. Here, we show that molecular dynamics simulations can be used to identify how this overall rigidification may be related to the allosteric communication within TCRs upon pMHC interaction via essential dynamics and nonbonded residue-residue interaction energy analyses. The residues taking part in the rigidification effect are highlighted with an intricate analysis on residue interaction changes, which lead to a detailed outline of the complex formation event. Our results indicate that residues of the Cβ domain of TCRs show significant differences in their nonbonded interactions upon complex formation. Moreover, the dynamic cross correlations between these residues are also increased, in line with their nonbonded interaction energy changes. Altogether, our approach may be valuable for elucidating intramolecular allosteric changes in the TCR structure upon pMHC interaction in molecular dynamics simulations.
Collapse
Affiliation(s)
- Elif Naz Bingöl
- Department of Bioengineering, Institute of Pure and Applied Sciences, Marmara University, 34722 Istanbul, Turkey
| | - Onur Serçinoğlu
- Department of Bioengineering, Faculty of Engineering, Gebze Technical University, Kocaeli 41400, Turkey
| | - Pemra Ozbek
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul 34722, Turkey
| |
Collapse
|
18
|
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: 0.8] [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
|
19
|
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: 1.0] [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
|
20
|
Hoffmann MM, Slansky JE. T-cell receptor affinity in the age of cancer immunotherapy. Mol Carcinog 2020; 59:862-870. [PMID: 32386086 DOI: 10.1002/mc.23212] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
The strength of the interaction between T-cell receptors (TCRs) and their ligands, peptide/major histocompatibility complex complexes (pMHCs), is one of the most frequently discussed and investigated features of T cells in immuno-oncology today. Although there are many molecules on the surface of T cells that interact with ligands on other cells, the TCR/pMHC is the only receptor-ligand pair that offers antigen specificity and dictates the functional response of the T cell. The strength of the TCR/pMHC interaction, along with the environment in which this interaction takes place, is key to how the T cell will respond. The TCR repertoire of T cells that interact with tumor-associated antigens is vast, although typically of low affinity. Here, we focus on the low-affinity interactions between TCRs from CD8+ T cells and different models used in immuno-oncology.
Collapse
Affiliation(s)
- Michele M Hoffmann
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jill E Slansky
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
21
|
Kristi N, Gafur A, Kong L, Ma X, Ye Z, Wang G. Atomic Force Microscopy in Mechanoimmunology Analysis: A New Perspective for Cancer Immunotherapy. Biotechnol J 2020; 15:e1900559. [PMID: 32240578 DOI: 10.1002/biot.201900559] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/08/2020] [Indexed: 01/05/2023]
Abstract
Immunotherapy has remarkable success outcomes against hematological malignancies with high rates of complete remission. To date, many studies have been conducted to increase its effectiveness in other types of cancer. However, it still yields unsatisfying results in solid tumor therapy. This limitation is partly attributed to the lack of understanding of how immunotherapy works in cancer from other perspectives. The traditional studies focus on the biological and chemical perspectives to determine which molecular substrates are involved in the immune system that can eradicate cancer cells. In the last decades, accumulating evidence has shown that physical properties also play important roles in the immune system to combat cancer, which is studied in mechanoimmunology. Mechanoimmunology analysis requires special tools; and herein, atomic force microscopy (AFM) appears as a versatile tool to determine and quantify the mechanical properties of a sample in nanometer precisions. Owing to its multifunctional capabilities, AFM can be used to explore immune system function from the physical perspective. This review paper explains the mechanoimmunology of how immune systems work through AFM, which includes mechanosignaling, mechanosensing, and mechanotransduction, with the aim to deepen the understanding of the mechanistic role of immunotherapy for further development in cancer treatment.
Collapse
Affiliation(s)
- Natalia Kristi
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, 400030, China
| | - Alidha Gafur
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, 400030, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Central Hospital of Chongqing University, Chongqing Emergency Medical Center, Chongqing, 400014, P. R. China
| | - Xinshuang Ma
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, 400030, China
| | - Zhiyi Ye
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, 400030, China
| |
Collapse
|
22
|
Kaitao L, William R, Zhou Y, Cheng Z. Single-molecule investigations of T-cell activation. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019; 12:102-110. [PMID: 32296738 PMCID: PMC7158867 DOI: 10.1016/j.cobme.2019.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
T-cell activation is the central event governing its development, differentiation, and effector functions. T-cell activation is initiated by the direct physical interaction of the T cell antigen receptor (TCR) with cognate peptide presented by the major histocompatibility complex (pMHC) molecule expressed on the antigen presenting cell (APC) surface. Since the identification of TCR as the only receptor for antigen on T cells three decades ago, studies have elucidated the major molecular players and signaling events responding to TCR stimulation. However, the question of how the physical event of pMHC binding is converted across the membrane into chemical events to initiate signal transduction remains elusive. Here we review recent investigations of T-cell activation using single-molecule force and fluorescence techniques that shed new light on this key question.
Collapse
Affiliation(s)
- Li Kaitao
- Wallace H. Coulter Department of Biomedical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Rittase William
- Wallace H. Coulter Department of Biomedical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Yuan Zhou
- George W. Woodruff School of Mechanical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Zhu Cheng
- Wallace H. Coulter Department of Biomedical Engineering
- George W. Woodruff School of Mechanical Engineering
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
23
|
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
|
24
|
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: 3.8] [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
|
25
|
Riley TP, Baker BM. The intersection of affinity and specificity in the development and optimization of T cell receptor based therapeutics. Semin Cell Dev Biol 2018; 84:30-41. [DOI: 10.1016/j.semcdb.2017.10.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 10/07/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022]
|
26
|
Li L, Hu J, Xu G, Song F. Binding constant of cell adhesion receptors and substrate-immobilized ligands depends on the distribution of ligands. Phys Rev E 2018; 97:012405. [PMID: 29448355 DOI: 10.1103/physreve.97.012405] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Indexed: 12/16/2022]
Abstract
Cell-cell adhesion and the adhesion of cells to tissues and extracellular matrix, which are pivotal for immune response, tissue development, and cell locomotion, depend sensitively on the binding constant of receptor and ligand molecules anchored on the apposing surfaces. An important question remains of whether the immobilization of ligands affects the affinity of binding with cell adhesion receptors. We have investigated the adhesion of multicomponent membranes to a flat substrate coated with immobile ligands using Monte Carlo simulations of a statistical mesoscopic model with biologically relevant parameters. We find that the binding of the adhesion receptors to ligands immobilized on the substrate is strongly affected by the ligand distribution. In the case of ligand clusters, the receptor-ligand binding constant can be significantly enhanced due to the less translational entropy loss of lipid-raft domains in the model cell membranes upon the formation of additional complexes. For ligands randomly or uniformly immobilized on the substrate, the binding constant is rather decreased since the receptors localized in lipid-raft domains have to pay an energetic penalty in order to bind ligands. Our findings help to understand why cell-substrate adhesion experiments for measuring the impact of lipid rafts on the receptor-ligand interactions led to contradictory results.
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
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China.,Shenzhen Institute of Research, Nanjing University, Shenzhen 518057, China
| | - Guangkui Xu
- School of Aerospace, Xi'an Jiaotong University, Xi'an 710049, China
| | - Fan Song
- 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.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
27
|
Spear TT, Wang Y, Smith TW, Simms PE, Garrett-Mayer E, Hellman LM, Baker BM, Nishimura MI. Altered Peptide Ligands Impact the Diversity of Polyfunctional Phenotypes in T Cell Receptor Gene-Modified T Cells. Mol Ther 2018; 26:996-1007. [PMID: 29503203 DOI: 10.1016/j.ymthe.2018.01.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/23/2022] Open
Abstract
The use of T cell receptor (TCR) gene-modified T cells in adoptive cell transfer has had promising clinical success, but often, simple preclinical evaluation does not necessarily accurately predict treatment efficacy or safety. Preclinical studies generally evaluate one or a limited number of type 1 cytokines to assess antigen recognition. However, recent studies have implicated other "typed" T cells in effective anti-tumor/viral immunity, and limited functional evaluations may underestimate cross-reactivity. In this study, we use an altered peptide ligand (APL) model and multi-dimensional flow cytometry to evaluate polyfunctionality of TCR gene-modified T cells. Evaluating six cytokines and the lytic marker CD107a on a per cell basis revealed remarkably diverse polyfunctional phenotypes within a single T cell culture and among peripheral blood lymphocyte (PBL) donors. This polyfunctional assessment identified unexpected phenotypes, including cells producing both type 1 and type 2 cytokines, and highlighted interferon γneg (IFNγneg) antigen-reactive populations overlooked in our previous studies. Additionally, APLs skewed functional phenotypes to be less polyfunctional, which was not necessarily related to changes in TCR-peptide-major histocompatibility complex (pMHC) affinity. A better understanding of gene-modified T cell functional diversity may help identify optimal therapeutic phenotypes, predict clinical responses, anticipate off-target recognition, and improve the design and delivery of TCR gene-modified T cells.
Collapse
Affiliation(s)
- Timothy T Spear
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153, USA.
| | - Yuan Wang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Thomas W Smith
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153, USA
| | - Patricia E Simms
- Flow Cytometry Core Facility, Office of Research Services, Loyola University Chicago, Maywood, IL, 60153 USA
| | - Elizabeth Garrett-Mayer
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29415, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29415, USA
| | - Lance M Hellman
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Brian M Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Michael I Nishimura
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
28
|
Allard M, Hebeisen M, Rufer N. Assessing T Cell Receptor Affinity and Avidity Against Tumor Antigens. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
29
|
Hickey JW, Vicente FP, Howard GP, Mao HQ, Schneck JP. Biologically Inspired Design of Nanoparticle Artificial Antigen-Presenting Cells for Immunomodulation. NANO LETTERS 2017; 17:7045-7054. [PMID: 28994285 PMCID: PMC6709596 DOI: 10.1021/acs.nanolett.7b03734] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Particles engineered to engage and interact with cell surface ligands and to modulate cells can be harnessed to explore basic biological questions as well as to devise cellular therapies. Biology has inspired the design of these particles, such as artificial antigen-presenting cells (aAPCs) for use in immunotherapy. While much has been learned about mimicking antigen presenting cell biology, as we decrease the size of aAPCs to the nanometer scale, we need to extend biomimetic design to include considerations of T cell biology-including T-cell receptor (TCR) organization. Here we describe the first quantitative analysis of particle size effect on aAPCs with both Signals 1 and 2 based on T cell biology. We show that aAPCs, larger than 300 nm, activate T cells more efficiently than smaller aAPCs, 50 nm. The 50 nm aAPCs require saturating doses or require artificial magnetic clustering to activate T cells. Increasing ligand density alone on the 50 nm aAPCs did not increase their ability to stimulate CD8+ T cells, confirming the size-dependent phenomenon. These data support the need for multireceptor ligation and activation of T-cell receptor (TCR) nanoclusters of similar sizes to 300 nm aAPCs. Quantitative analysis and modeling of a nanoparticle system provides insight into engineering constraints of aAPCs for T cell immunotherapy applications and offers a case study for other cell-modulating particles.
Collapse
Affiliation(s)
- John W. Hickey
- Department of Biomedical Engineering, School of Medicine
- Institute for Cell Engineering, School of Medicine
- Translational Tissue Engineering Center
- Institute for Nanobiotechnology
| | | | - Gregory P. Howard
- Department of Biomedical Engineering, School of Medicine
- Institute for Nanobiotechnology
| | - Hai-Quan Mao
- Translational Tissue Engineering Center
- Institute for Nanobiotechnology
- Department of Materials Science and Engineering, Whiting School of Engineering
| | - Jonathan P. Schneck
- Institute for Cell Engineering, School of Medicine
- Department of Pathology, School of Medicine
- Institute for Nanobiotechnology
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Corresponding Author: . Phone: 410-614-4589
| |
Collapse
|
30
|
Singh NK, Riley TP, Baker SCB, Borrman T, Weng Z, Baker BM. Emerging Concepts in TCR Specificity: Rationalizing and (Maybe) Predicting Outcomes. THE JOURNAL OF IMMUNOLOGY 2017; 199:2203-2213. [PMID: 28923982 DOI: 10.4049/jimmunol.1700744] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
T cell specificity emerges from a myriad of processes, ranging from the biological pathways that control T cell signaling to the structural and physical mechanisms that influence how TCRs bind peptides and MHC proteins. Of these processes, the binding specificity of the TCR is a key component. However, TCR specificity is enigmatic: TCRs are at once specific but also cross-reactive. Although long appreciated, this duality continues to puzzle immunologists and has implications for the development of TCR-based therapeutics. In this review, we discuss TCR specificity, emphasizing results that have emerged from structural and physical studies of TCR binding. We show how the TCR specificity/cross-reactivity duality can be rationalized from structural and biophysical principles. There is excellent agreement between predictions from these principles and classic predictions about the scope of TCR cross-reactivity. We demonstrate how these same principles can also explain amino acid preferences in immunogenic epitopes and highlight opportunities for structural considerations in predictive immunology.
Collapse
Affiliation(s)
- Nishant K Singh
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556; and
| | - Timothy P Riley
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556; and
| | - Sarah Catherine B Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556; and
| | - Tyler Borrman
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Brian M Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556; .,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556; and
| |
Collapse
|
31
|
Chen Y, Ju L, Rushdi M, Ge C, Zhu C. Receptor-mediated cell mechanosensing. Mol Biol Cell 2017; 28:3134-3155. [PMID: 28954860 PMCID: PMC5687017 DOI: 10.1091/mbc.e17-04-0228] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 09/06/2017] [Accepted: 09/19/2017] [Indexed: 12/22/2022] Open
Abstract
Mechanosensing depicts the ability of a cell to sense mechanical cues, which under some circumstances is mediated by the surface receptors. In this review, a four-step model is described for receptor-mediated mechanosensing. Platelet GPIb, T-cell receptor, and integrins are used as examples to illustrate the key concepts and players in this process. Mechanosensing describes the ability of a cell to sense mechanical cues of its microenvironment, including not only all components of force, stress, and strain but also substrate rigidity, topology, and adhesiveness. This ability is crucial for the cell to respond to the surrounding mechanical cues and adapt to the changing environment. Examples of responses and adaptation include (de)activation, proliferation/apoptosis, and (de)differentiation. Receptor-mediated cell mechanosensing is a multistep process that is initiated by binding of cell surface receptors to their ligands on the extracellular matrix or the surface of adjacent cells. Mechanical cues are presented by the ligand and received by the receptor at the binding interface; but their transmission over space and time and their conversion into biochemical signals may involve other domains and additional molecules. In this review, a four-step model is described for the receptor-mediated cell mechanosensing process. Platelet glycoprotein Ib, T-cell receptor, and integrins are used as examples to illustrate the key concepts and players in this process.
Collapse
Affiliation(s)
- Yunfeng Chen
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Lining Ju
- Charles Perkins Centre and Heart Research Institute, University of Sydney, Camperdown, NSW 2006, Australia
| | - Muaz Rushdi
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332.,Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Chenghao Ge
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332.,Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Cheng Zhu
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332 .,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332.,Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
32
|
Catch Bonds at T Cell Interfaces: Impact of Surface Reorganization and Membrane Fluctuations. Biophys J 2017; 113:120-131. [PMID: 28700910 DOI: 10.1016/j.bpj.2017.05.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/07/2017] [Accepted: 05/19/2017] [Indexed: 01/30/2023] Open
Abstract
Catch bonds are characterized by average lifetimes that initially increase with increasing tensile force. Recently, they have been implicated in T cell activation, where small numbers of antigenic receptor-ligand bonds at a cell-cell interface can stimulate a T cell. Here, we use computational methods to investigate small numbers of bonds at the interface between two membranes. We characterize the time-dependent forces on the bonds in response to changes in the membrane shape and the organization of other surface molecules. We then determine the distributions of bond lifetimes using recent force-dependent lifetime data for T cell receptors bound to various ligands. Strong agonists, which exhibit catch bond behavior, are markedly more likely to remain intact than an antagonist whose average lifetime decreases with increasing force. Thermal fluctuations of the membrane shape enhance the decay of the average force on a bond, but also lead to fluctuations of the force. These fluctuations promote bond rupture, but the effect is buffered by catch bonds. When more than one bond is present, the bonds experience reduced average forces that depend on their relative positions, leading to changes in bond lifetimes. Our results highlight the importance of force-dependent binding kinetics when bonds experience time-dependent and fluctuating forces, as well as potential consequences of collective bond behavior relevant to T cell activation.
Collapse
|
33
|
Williams CM, Schonnesen AA, Zhang SQ, Ma KY, He C, Yamamoto T, Eckhardt SG, Klebanoff CA, Jiang N. Normalized Synergy Predicts That CD8 Co-Receptor Contribution to T Cell Receptor (TCR) and pMHC Binding Decreases As TCR Affinity Increases in Human Viral-Specific T Cells. Front Immunol 2017; 8:894. [PMID: 28804489 PMCID: PMC5532383 DOI: 10.3389/fimmu.2017.00894] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 07/12/2017] [Indexed: 11/13/2022] Open
Abstract
The discovery of naturally occurring T cell receptors (TCRs) that confer specific, high-affinity recognition of pathogen and cancer-associated antigens remains a major goal in cellular immunotherapies. The contribution of the CD8 co-receptor to the interaction between the TCR and peptide-bound major histocompatibility complex (pMHC) has previously been correlated with the activation and responsiveness of CD8+ T cells. However, these studies have been limited to model systems of genetically engineered hybridoma TCRs or transgenic mouse TCRs against either a single epitope or an array of altered peptide ligands. CD8 contribution in a native human antigen-specific T cell response remains elusive. Here, using Hepatitis C Virus-specific precursor CTLs spanning a large range of TCR affinities, we discovered that the functional responsiveness of any given TCR correlated with the contribution of CD8 to TCR/pMHC binding. Furthermore, we found that CD8 contribution to TCR/pMHC binding in the two-dimensional (2D) system was more accurately reflected by normalized synergy (CD8 cooperation normalized by total TCR/pMHC bonds) rather than synergy (total CD8 cooperation) alone. While synergy showed an increasing trend with TCR affinity, normalized synergy was demonstrated to decrease with the increase of TCR affinity. Critically, normalized synergy was shown to correlate with CTL functionality and peptide sensitivity, corroborating three-dimensional (3D) analysis of CD8 contribution with respect to TCR affinity. In addition, we identified TCRs that were independent of CD8 for TCR/pMHC binding. Our results resolve the current discrepancy between 2D and 3D analysis on CD8 contribution to TCR/pMHC binding, and demonstrate that naturally occurring high-affinity TCRs are more capable of CD8-independent interactions that yield greater functional responsiveness even with CD8 blocking. Taken together, our data suggest that addition of the normalized synergy parameter to our previously established TCR discovery platform using 2D TCR affinity and sequence test would allow for selection of TCRs specific to any given antigen with the desirable attributes of high TCR affinity, CD8 co-receptor independence and functional superiority. Utilizing TCRs with less CD8 contribution could be beneficial for adoptive cell transfer immunotherapies using naturally occurring or genetically engineered T cells against viral or cancer-associated antigens.
Collapse
Affiliation(s)
- Chad M Williams
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Alexandra A Schonnesen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Shu-Qi Zhang
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Ke-Yue Ma
- Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX, United States
| | - Chenfeng He
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Tori Yamamoto
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, United States
| | - S Gail Eckhardt
- LIVESTRONG Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Christopher A Klebanoff
- Center for Cell Engineering, Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, United States.,Parker Institute for Cancer Immunotherapy, MSKCC, New York, NY, United States
| | - Ning Jiang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States.,Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
34
|
Allard M, Couturaud B, Carretero-Iglesia L, Duong MN, Schmidt J, Monnot GC, Romero P, Speiser DE, Hebeisen M, Rufer N. TCR-ligand dissociation rate is a robust and stable biomarker of CD8+ T cell potency. JCI Insight 2017; 2:92570. [PMID: 28724801 DOI: 10.1172/jci.insight.92570] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/15/2017] [Indexed: 12/16/2022] Open
Abstract
Despite influencing many aspects of T cell biology, the kinetics of T cell receptor (TCR) binding to peptide-major histocompatibility molecules (pMHC) remain infrequently determined in patient monitoring or for adoptive T cell therapy. Using specifically designed reversible fluorescent pMHC multimeric complexes, we performed a comprehensive study of TCR-pMHC off-rates combined with various functional assays on large libraries of self/tumor- and virus-specific CD8+ T cell clones from melanoma patients and healthy donors. We demonstrate that monomeric TCR-pMHC dissociation rates accurately predict the extent of cytotoxicity, cytokine production, polyfunctionality, cell proliferation, activating/inhibitory receptor expression, and in vivo antitumor potency of naturally occurring antigen-specific CD8+ T cells. Our data also confirm the superior binding avidities of virus-specific T cells as compared with self/tumor-specific T cell clonotypes (n > 300). Importantly, the TCR-pMHC off-rate is a more stable and robust biomarker of CD8+ T cell potency than the frequently used functional assays/metrics that depend on the T cell's activation state, and therefore show major intra- and interexperimental variability. Taken together, our data show that the monomeric TCR-pMHC off-rate is highly useful for the ex vivo high-throughput functional assessment of antigen-specific CD8+ T cell responses and a strong candidate as a biomarker of T cell therapeutic efficacy.
Collapse
Affiliation(s)
- Mathilde Allard
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Barbara Couturaud
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Laura Carretero-Iglesia
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Minh Ngoc Duong
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Julien Schmidt
- Ludwig Cancer Research, University of Lausanne, Epalinges, Switzerland
| | | | - Pedro Romero
- Ludwig Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Epalinges, Switzerland.,Ludwig Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Michael Hebeisen
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Nathalie Rufer
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Epalinges, Switzerland.,Ludwig Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
35
|
Li L, Hu J, Shi X, Shao Y, Song F. Lipid rafts enhance the binding constant of membrane-anchored receptors and ligands. SOFT MATTER 2017; 13:4294-4304. [PMID: 28573272 DOI: 10.1039/c7sm00572e] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Gaining insights into the binding of membrane-anchored receptors and ligands that mediate cell adhesion and signal transduction is of great significance for understanding numerous physiological processes driven by intercellular communication. Lipid rafts, microdomains in cell membranes enriched in cholesterol and saturated lipids such as sphingomyelin, are believed to serve as the essential platforms to recruit protein molecules for biological functions. An important question remains how the lipid rafts affect the binding constant of membrane-anchored receptors and ligands. We have investigated the adhesion of multicomponent membranes by using Monte Carlo simulations of a mesoscopic model with biologically relevant parameters. We find that the preferential partitioning of membrane-anchored receptor and ligand proteins in the lipid rafts significantly increases the binding constant of those proteins, in cooperation with the shape fluctuations of the membranes caused by thermal excitations. The binding constant can even be greater than that of the same receptors and ligands anchored to two apposing supported, planar membranes without shape fluctuations. The membrane shape fluctuations facilitate the binding of the anchored receptors and ligands, in contrast to the case of homogeneous membranes. Our results suggest that cells might regulate the binding of membrane-anchored receptor and ligand proteins by modulating the properties of lipid rafts such as area fraction, size and the affinity of rafts to the proteins.
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
|
36
|
Abstract
Upon engagement with a specific ligand, a cell surface receptor transduces intracellular signals to activate various cellular functions. This chapter describes a set of biomechanical methods for analyzing the characteristics of cross-junctional receptor-ligand interactions at the surface of living cells. These methods combine the characterization of kinetics of receptor-ligand binding with real-time imaging of intracellular calcium fluxes, which allow researchers to assess how the signal initiated from single receptor-ligand engagement is transduced across the cell membrane. A major application of these methods is the analysis of antigen recognition by triggering of the T cell receptor (TCR). Three related methods are described in this chapter: (1) the micropipette adhesion assay, (2) the biomembrane force probe (BFP) assay, and (3) combining BFP with fluorescence microscopy (fBFP). In all cases, an ultrasoft human red blood cell (RBC) is used as an ultrasensitive mechanical force probe. The micropipette assay detects binding events visually. The BFP uses a high-speed camera and real-time image tracking techniques to measure mechanical variables on a single molecular bond with up to ~1 pN (10-12 Newton), ~3 nm (10-9 m), and ~0.5 ms (10-3 s) in force, spatial, and temporal resolution, respectively. As an upgrade to the BFP, the fBFP simultaneously images binding-triggered intracellular calcium signals on a single live cell. These technologies can be widely used to study other membrane receptor-ligand interactions and signaling under mechanical regulation.
Collapse
|
37
|
Hwang I, Kim K, Choi S, Lomunova M. Potentiation of T Cell Stimulatory Activity by Chemical Fixation of a Weak Peptide-MHC Complex. Mol Cells 2017; 40:24-36. [PMID: 28152301 PMCID: PMC5303886 DOI: 10.14348/molcells.2017.2218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/12/2016] [Accepted: 12/27/2016] [Indexed: 12/13/2022] Open
Abstract
The stability of peptide-MHC complex (pMHC) is an important factor to shape the fate of peptide-specific T cell immune response, but how it influences on T cell activation process is poorly understood. To better understand that, we investigated various T cell activation events driven by Ld MHCI loaded with graded concentrations of P2Ca and QL9 peptides, respectively, with 2C TCR Tg T cells; the binding strength of P2Ca for Ld is measurably weaker than that of QL9, but either peptides in the context of Ld interact with 2C TCR with a similar strength. When their concentrations required for early T cell activation events, which occur within several minutes to an hour, were concerned, EC50s of QL9 were about 100 folds lower than those of P2Ca, which was expected from their association constants for Ld. When EC50s for late activation events, which takes over several hours to occur, were concerned, the differences grew even larger (> 300 folds), suggesting that, due to weak binding, Ld/P2Ca dissociate from each other more easily to lose its antigenicity in a short time. Accordingly, fixation of Ld/P2Ca with paraformaldehyde resulted in a significant improvement in its immunogenicity. These results imply that binding strength of a peptide for a MHC is a critical factor to determine the duration of pMHC-mediated T cell activation and thus the attainment of productive T cell activation. It is also suggested that paraformaldehyde fixation should be an effective tool to ameliorate the immunogenicity of pMHC with a poor stability.
Collapse
Affiliation(s)
- Inkyu Hwang
- Department of Chemistry and Chemical Biology, The Scripps Research Institute, La Jolla, California,
USA
- College of Pharmacy, Chungnam National University, Daejeon 34134,
Korea
| | - Kwangmi Kim
- Department of Chemistry and Chemical Biology, The Scripps Research Institute, La Jolla, California,
USA
- College of Pharmacy, Dankook University, Yongin 16890,
Korea
| | - Sojin Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134,
Korea
| | - Maria Lomunova
- College of Pharmacy, Chungnam National University, Daejeon 34134,
Korea
| |
Collapse
|
38
|
Zhang H, Lim HS, Knapp B, Deane CM, Aleksic M, Dushek O, van der Merwe PA. The contribution of major histocompatibility complex contacts to the affinity and kinetics of T cell receptor binding. Sci Rep 2016; 6:35326. [PMID: 27734930 PMCID: PMC5062128 DOI: 10.1038/srep35326] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/28/2016] [Indexed: 11/09/2022] Open
Abstract
The interaction between the T cell antigen receptor (TCR) and antigenic peptide in complex with major histocompatibility complex (MHC) molecules is a crucial step in T cell activation. The relative contributions of TCR:peptide and TCR:MHC contacts to the overall binding energy remain unclear. This has important implications for our understanding of T cell development and function. In this study we used site directed mutagenesis to estimate the contribution of HLA-A2 side-chains to the binding of four TCRs. Our results show that these TCRs have very different energetic ‘footprints’ on HLA-A2, with no residues contributing to all TCR interactions. The estimated overall contribution of MHC side-chains to the total interaction energy was variable, with lower limits ranging from 11% to 50%. Kinetic analysis suggested a minor and variable contribution of MHC side-chains to the transition state complex, arguing against a two-step mechanism for TCR binding.
Collapse
Affiliation(s)
- Hao Zhang
- Sir William Dunn School of Pathology, University of Oxford, United Kingdom
| | - Hong-Sheng Lim
- Sir William Dunn School of Pathology, University of Oxford, United Kingdom
| | - Berhard Knapp
- Department of Statistics, University of Oxford, United Kingdom
| | | | - Milos Aleksic
- Sir William Dunn School of Pathology, University of Oxford, United Kingdom
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, United Kingdom
| | | |
Collapse
|
39
|
Xu GK, Qian J, Hu J. The glycocalyx promotes cooperative binding and clustering of adhesion receptors. SOFT MATTER 2016; 12:4572-4583. [PMID: 27102288 DOI: 10.1039/c5sm03139g] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Cell adhesion plays a pivotal role in various biological processes, e.g., immune responses, cancer metastasis, and stem cell differentiation. The adhesion behaviors depend subtly on the binding kinetics of receptors and ligands restricted at the cell-substrate interfaces. Although much effort has been directed toward investigating the kinetics of adhesion molecules, the role of the glycocalyx, anchored on cell surfaces as an exterior layer, is still unclear. In this paper, we propose a theoretical approach to study the collective binding kinetics of a few and a large number of binders in the presence of the glycocalyx, representing the cases of initial and mature adhesions of cells, respectively. The analytical results are validated by finding good agreement with our Monte Carlo simulations. In the force loading case, the on-rate and affinity increase as more bonds form, whereas this cooperative effect is not observed in the displacement loading case. The increased thickness and stiffness of the glycocalyx tend to decrease the affinity for a few bonds, while they have less influence on the affinity for a large number of bonds. Moreover, for a flexible membrane with thermally-excited shape fluctuations, the glycocalyx is exhibited to promote the formation of bond clusters, mainly due to the cooperative binding of binders. This study helps to understand the cooperative kinetics of adhesion receptors under physiologically relevant loading conditions and sheds light on the novel role of the glycocalyx in cell adhesion.
Collapse
Affiliation(s)
- Guang-Kui Xu
- International Center for Applied Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Jin Qian
- Department of Engineering Mechanics, Soft Matter Research Center, Zhejiang University, Hangzhou 310027, China
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
| |
Collapse
|
40
|
Xu GK, Liu Z, Feng XQ, Gao H. Tension-compression asymmetry in the binding affinity of membrane-anchored receptors and ligands. Phys Rev E 2016; 93:032411. [PMID: 27078394 DOI: 10.1103/physreve.93.032411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Indexed: 06/05/2023]
Abstract
Cell adhesion plays a crucial role in many biological processes of cells, e.g., immune responses, tissue morphogenesis, and stem cell differentiation. An essential problem in the molecular mechanism of cell adhesion is to characterize the binding affinity of membrane-anchored receptors and ligands under different physiological conditions. In this paper, a theoretical model is presented to study the binding affinity between a large number of anchored receptors and ligands under both tensile and compressive stresses, and corroborated by demonstrating excellent agreement with Monte Carlo simulations. It is shown that the binding affinity becomes lower as the magnitude of the applied stress increases, and drops to zero at a critical tensile or compressive stress. Interestingly, the critical compressive stress is found to be substantially smaller than the critical tensile stress for relatively long and flexible receptor-ligand complexes. This counterintuitive finding is explained by using the Euler instability theory of slender columns under compression. The tension-compression asymmetry in the binding affinity of anchored receptors and ligands depends subtly on the competition between the breaking and instability of their complexes. This study helps in understanding the role of mechanical forces in cell adhesion mediated by specific binding molecules.
Collapse
Affiliation(s)
- Guang-Kui Xu
- International Center for Applied Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zishun Liu
- International Center for Applied Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Huajian Gao
- School of Engineering, Brown University, Providence, Rhode Island 02912, USA
| |
Collapse
|
41
|
Ju L, Qian J, Zhu C. Transport regulation of two-dimensional receptor-ligand association. Biophys J 2016; 108:1773-1784. [PMID: 25863068 DOI: 10.1016/j.bpj.2015.02.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/15/2015] [Accepted: 02/24/2015] [Indexed: 01/12/2023] Open
Abstract
The impact of flow disturbances on platelet adhesion is complex and incompletely understood. At the molecular scale, platelet glycoprotein Ibα (GPIbα) must associate with the von Willebrand factor A1 domain (VWF-A1) with a rapid on-rate under high hemodynamic forces, as occurs in arterial thrombosis, where various transport mechanisms are at work. Here, we theoretically modeled the coupled transport-reaction process of the two-dimensional (2D) receptor-ligand association kinetics in a biomembrane force probe to explicitly account for the effects of molecular length, confinement stiffness, medium viscosity, surface curvature, and separation distance. We experimentally verified the theoretical approach by visualizing association and dissociation of individual VWF-A1-GPIbα bonds in a real-time thermal fluctuation assay. The apparent on-rate, reciprocal of the average time intervals between sequential bonds, decreased with the increasing gap distance between A1- and GPIbα-bearing surfaces with an 80-nm threshold (beyond which bond formation became prohibitive) identified as the combined contour length of the receptor and ligand molecules. The biomembrane force probe spring constant and diffusivity of the protein-bearing beads also significantly influenced the apparent on-rate, in accordance with the proposed transport mechanisms. The global agreement between the experimental data and the model predictions supports the hypothesis that receptor-ligand association behaves distinctly in the transport- and reaction-limited scenarios. To our knowledge, our results represent the first detailed quantification of physical regulation of the 2D on-rate that allows platelets to sense and respond to local changes in their hemodynamic environment. In addition, they provide an approach for determining the intrinsic kinetic parameters that employs simultaneous experimental measurements and theoretical modeling of bond association in a single assay. The 2D intrinsic forward rate for VWF-A1-GPIbα association was determined from the measurements to be (3.5 ± 0.67) × 10(-4)μm(2) s(-1).
Collapse
Affiliation(s)
- Lining Ju
- Coulter School of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia; Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Jin Qian
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Cheng Zhu
- Coulter School of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia; Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia.
| |
Collapse
|
42
|
Hebeisen M, Allard M, Gannon PO, Schmidt J, Speiser DE, Rufer N. Identifying Individual T Cell Receptors of Optimal Avidity for Tumor Antigens. Front Immunol 2015; 6:582. [PMID: 26635796 PMCID: PMC4649060 DOI: 10.3389/fimmu.2015.00582] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/30/2015] [Indexed: 02/02/2023] Open
Abstract
Cytotoxic T cells recognize, via their T cell receptors (TCRs), small antigenic peptides presented by the major histocompatibility complex (pMHC) on the surface of professional antigen-presenting cells and infected or malignant cells. The efficiency of T cell triggering critically depends on TCR binding to cognate pMHC, i.e., the TCR–pMHC structural avidity. The binding and kinetic attributes of this interaction are key parameters for protective T cell-mediated immunity, with stronger TCR–pMHC interactions conferring superior T cell activation and responsiveness than weaker ones. However, high-avidity TCRs are not always available, particularly among self/tumor antigen-specific T cells, most of which are eliminated by central and peripheral deletion mechanisms. Consequently, systematic assessment of T cell avidity can greatly help distinguishing protective from non-protective T cells. Here, we review novel strategies to assess TCR–pMHC interaction kinetics, enabling the identification of the functionally most-relevant T cells. We also discuss the significance of these technologies in determining which cells within a naturally occurring polyclonal tumor-specific T cell response would offer the best clinical benefit for use in adoptive therapies, with or without T cell engineering.
Collapse
Affiliation(s)
- Michael Hebeisen
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Mathilde Allard
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Philippe O Gannon
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Julien Schmidt
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland ; TCMetrix Sàrl , Epalinges , Switzerland
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland ; Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland
| | - Nathalie Rufer
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland ; Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland
| |
Collapse
|
43
|
Martinez RJ, Evavold BD. Lower Affinity T Cells are Critical Components and Active Participants of the Immune Response. Front Immunol 2015; 6:468. [PMID: 26441973 PMCID: PMC4564719 DOI: 10.3389/fimmu.2015.00468] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 08/28/2015] [Indexed: 11/13/2022] Open
Abstract
Kinetic and biophysical parameters of T cell receptor (TCR) and peptide:MHC (pMHC) interaction define intrinsic factors required for T cell activation and differentiation. Although receptor ligand kinetics are somewhat cumbersome to assess experimentally, TCR:pMHC affinity has been shown to predict peripheral T cell functionality and potential for forming memory. Multimeric forms of pMHC monomers have often been used to provide an indirect readout of higher affinity T cells due to their availability and ease of use while allowing simultaneous definition of other functional and phenotypic characteristics. However, multimeric pMHC reagents have introduced a bias that underestimates the lower affinity components contained in the highly diverse TCR repertoires of all polyclonal T cell responses. Advances in the identification of lower affinity cells have led to the examination of these cells and their contribution to the immune response. In this review, we discuss the identification of high- vs. low-affinity T cells as well as their attributed signaling and functional differences. Lastly, mechanisms are discussed that maintain a diverse range of low- and high-affinity T cells.
Collapse
Affiliation(s)
- Ryan J. Martinez
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Brian D. Evavold
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| |
Collapse
|
44
|
Hong J, Persaud SP, Horvath S, Allen PM, Evavold BD, Zhu C. Force-Regulated In Situ TCR-Peptide-Bound MHC Class II Kinetics Determine Functions of CD4+ T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:3557-64. [PMID: 26336148 DOI: 10.4049/jimmunol.1501407] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/04/2015] [Indexed: 12/27/2022]
Abstract
We have recently shown that two-dimensional (2D) and force-regulated kinetics of TCR-peptide-bound MHC class I (pMHC-I) interactions predict responses of CD8(+) T cells. To test whether these findings are applicable to CD4(+) T cells, we analyzed the in situ 3.L2 TCR-pMHC-II interactions for a well-characterized panel of altered peptide ligands on the T cell surface using the adhesion frequency assay with a micropipette and the thermal fluctuation and force-clamp assays with a biomembrane force probe. We found that the 2D effective TCR-pMHC-II affinity and off-rate correlate with, but better predict the T cell response than, the corresponding measurements with the surface plasmon resonance in three dimensions. The 2D affinity of the CD4 for MHC-II was very low, approaching the detection limit, making it one to two orders of magnitude lower than the affinity of CD8 for MHC-I. In addition, the signal-dependent cooperation between TCR and coreceptor for pMHC binding previously observed for CD8 was not observed for CD4. Interestingly, force elicited TCR-pMHC-II catch-slip bonds for agonists but slip-only bonds for antagonists, thereby amplifying the power of discrimination between altered peptide ligands. These results show that the force-regulated 2D binding kinetics of the 3.L2 TCR for pMHC-II determine functions of CD4(+) T cells.
Collapse
Affiliation(s)
- Jinsung Hong
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332; Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Stephen P Persaud
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen Horvath
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Paul M Allen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Brian D Evavold
- Department of Immunology and Microbiology, Emory University School of Medicine, Atlanta, GA 30332; and
| | - Cheng Zhu
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332; Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332; Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
45
|
Chen Y, Liu B, Ju L, Hong J, Ji Q, Chen W, Zhu C. Fluorescence Biomembrane Force Probe: Concurrent Quantitation of Receptor-ligand Kinetics and Binding-induced Intracellular Signaling on a Single Cell. J Vis Exp 2015:e52975. [PMID: 26274371 DOI: 10.3791/52975] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Membrane receptor-ligand interactions mediate many cellular functions. Binding kinetics and downstream signaling triggered by these molecular interactions are likely affected by the mechanical environment in which binding and signaling take place. A recent study demonstrated that mechanical force can regulate antigen recognition by and triggering of the T-cell receptor (TCR). This was made possible by a new technology we developed and termed fluorescence biomembrane force probe (fBFP), which combines single-molecule force spectroscopy with fluorescence microscopy. Using an ultra-soft human red blood cell as the sensitive force sensor, a high-speed camera and real-time imaging tracking techniques, the fBFP is of ~1 pN (10(-12) N), ~3 nm and ~0.5 msec in force, spatial and temporal resolution. With the fBFP, one can precisely measure single receptor-ligand binding kinetics under force regulation and simultaneously image binding-triggered intracellular calcium signaling on a single live cell. This new technology can be used to study other membrane receptor-ligand interaction and signaling in other cells under mechanical regulation.
Collapse
Affiliation(s)
- Yunfeng Chen
- Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology
| | - Baoyu Liu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology
| | - Lining Ju
- Charles Perkins Centre, The University of Sydney
| | - Jinsung Hong
- Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology
| | - Qinghua Ji
- Institute of Biophysics, Laboratory of RNA Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences
| | - Wei Chen
- School of Medicine and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University
| | - Cheng Zhu
- Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology;
| |
Collapse
|
46
|
Liu B, Chen W, Natarajan K, Li Z, Margulies DH, Zhu C. The cellular environment regulates in situ kinetics of T-cell receptor interaction with peptide major histocompatibility complex. Eur J Immunol 2015; 45:2099-110. [PMID: 25944482 DOI: 10.1002/eji.201445358] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/17/2015] [Accepted: 04/30/2015] [Indexed: 11/12/2022]
Abstract
T cells recognize antigens at the two-dimensional (2D) interface with antigen-presenting cells (APCs), which trigger T-cell effector functions. T-cell functional outcomes correlate with 2D kinetics of membrane-embedded T-cell receptors (TCRs) binding to surface-tethered peptide-major histocompatibility complex molecules (pMHCs). However, most studies have measured TCR-pMHC kinetics for recombinant TCRs in 3D by surface plasmon resonance, which differs drastically from 2D measurements. Here, we compared pMHC dissociation from native TCR on the T-cell surface to recombinant TCR immobilized on glass surface or in solution. Force on TCR-pMHC bonds regulated their lifetimes differently for native than recombinant TCRs. Perturbing the cellular environment suppressed 2D on-rates but had no effect on 2D off-rate regardless of whether force was applied. In contrast, for the TCR interacting with its monoclonal antibody, the 2D on-rate was insensitive to cellular perturbations and the force-dependent off-rates were indistinguishable for native and recombinant TCRs. These data present novel features of TCR-pMHC kinetics that are regulated by the cellular environment, underscoring the limitations of 3D kinetics in predicting T-cell functions and calling for further elucidation of the underlying molecular and cellular mechanisms that regulate 2D kinetics in physiological settings.
Collapse
Affiliation(s)
- Baoyu Liu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Wei Chen
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kannan Natarajan
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Zhenhai Li
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - David H Margulies
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
47
|
Gannon PO, Wieckowski S, Baumgaertner P, Hebeisen M, Allard M, Speiser DE, Rufer N. Quantitative TCR:pMHC Dissociation Rate Assessment by NTAmers Reveals Antimelanoma T Cell Repertoires Enriched for High Functional Competence. THE JOURNAL OF IMMUNOLOGY 2015; 195:356-66. [PMID: 26002978 DOI: 10.4049/jimmunol.1403145] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/28/2015] [Indexed: 12/19/2022]
Abstract
Experimental models demonstrated that therapeutic induction of CD8 T cell responses may offer protection against tumors or infectious diseases providing that T cells have sufficiently high TCR/CD8:pMHC avidity for efficient Ag recognition and consequently strong immune functions. However, comprehensive characterization of TCR/CD8:pMHC avidity in clinically relevant situations has remained elusive. In this study, using the novel NTA-His tag-containing multimer technology, we quantified the TCR:pMHC dissociation rates (koff) of tumor-specific vaccine-induced CD8 T cell clones (n = 139) derived from seven melanoma patients vaccinated with IFA, CpG, and the native/EAA or analog/ELA Melan-A(MART-1)(26-35) peptide, binding with low or high affinity to MHC, respectively. We observed substantial correlations between koff and Ca(2+) mobilization (p = 0.016) and target cell recognition (p < 0.0001), with the latter independently of the T cell differentiation state. Our strategy was successful in demonstrating that the type of peptide impacted on TCR/CD8:pMHC avidity, as tumor-reactive T cell clones derived from patients vaccinated with the low-affinity (native) peptide expressed slower koff rates than those derived from patients vaccinated with the high-affinity (analog) peptide (p < 0.0001). Furthermore, we observed that the low-affinity peptide promoted the selective differentiation of tumor-specific T cells bearing TCRs with high TCR/CD8:pMHC avidity (p < 0.0001). Altogether, TCR:pMHC interaction kinetics correlated strongly with T cell functions. Our study demonstrates the feasibility and usefulness of TCR/CD8:pMHC avidity assessment by NTA-His tag-containing multimers of naturally occurring polyclonal T cell responses, which represents a strong asset for the development of immunotherapy.
Collapse
Affiliation(s)
- Philippe O Gannon
- Department of Oncology, Lausanne University Hospital and University of Lausanne, CH-1011 Lausanne, Switzerland; and
| | - Sébastien Wieckowski
- Department of Oncology, Lausanne University Hospital and University of Lausanne, CH-1011 Lausanne, Switzerland; and
| | - Petra Baumgaertner
- Ludwig Center for Cancer Research, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Michaël Hebeisen
- Department of Oncology, Lausanne University Hospital and University of Lausanne, CH-1011 Lausanne, Switzerland; and
| | - Mathilde Allard
- Department of Oncology, Lausanne University Hospital and University of Lausanne, CH-1011 Lausanne, Switzerland; and
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital and University of Lausanne, CH-1011 Lausanne, Switzerland; and Ludwig Center for Cancer Research, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Nathalie Rufer
- Department of Oncology, Lausanne University Hospital and University of Lausanne, CH-1011 Lausanne, Switzerland; and Ludwig Center for Cancer Research, University of Lausanne, CH-1011 Lausanne, Switzerland
| |
Collapse
|
48
|
Abstract
Molecular force spectroscopy has become a powerful tool to study how mechanics regulates biology, especially the mechanical regulation of molecular interactions and its impact on cellular functions. This force-driven methodology has uncovered a wealth of new information of the physical chemistry of molecular bonds for various biological systems. The new concepts, qualitative and quantitative measures describing bond behavior under force, and structural bases underlying these phenomena have substantially advanced our fundamental understanding of the inner workings of biological systems from the nanoscale (molecule) to the microscale (cell), elucidated basic molecular mechanisms of a wide range of important biological processes, and provided opportunities for engineering applications. Here, we review major force spectroscopic assays, conceptual developments of mechanically regulated kinetics of molecular interactions, and their biological relevance. We also present current challenges and highlight future directions.
Collapse
Affiliation(s)
- Baoyu Liu
- Coulter Department of Biomedical Engineering
| | | | | |
Collapse
|
49
|
Perica K, Kosmides AK, Schneck JP. Linking form to function: Biophysical aspects of artificial antigen presenting cell design. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:781-90. [PMID: 25200637 PMCID: PMC4344884 DOI: 10.1016/j.bbamcr.2014.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/15/2014] [Accepted: 09/01/2014] [Indexed: 12/22/2022]
Abstract
Artificial antigen presenting cells (aAPCs) are engineered platforms for T cell activation and expansion, synthesized by coupling T cell activating proteins to the surface of cell lines or biocompatible particles. They can serve both as model systems to study the basic aspects of T cell signaling and translationally as novel approaches for either active or adoptive immunotherapy. Historically, these reductionist systems have not been designed to mimic the temporally and spatially complex interactions observed during endogenous T cell-APC contact, which include receptor organization at both micro- and nanoscales and dynamic changes in cell and membrane morphologies. Here, we review how particle size and shape, as well as heterogenous distribution of T cell activating proteins on the particle surface, are critical aspects of aAPC design. In doing so, we demonstrate how insights derived from endogenous T cell activation can be applied to optimize aAPC, and in turn how aAPC platforms can be used to better understand endogenous T cell stimulation. This article is part of a Special Issue entitled: Nanoscale membrane organisation and signalling.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan P Schneck
- Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
50
|
Klotzsch E, Stiegler J, Ben-Ishay E, Gaus K. Do mechanical forces contribute to nanoscale membrane organisation in T cells? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:822-9. [PMID: 25447546 DOI: 10.1016/j.bbamcr.2014.10.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/26/2014] [Accepted: 10/25/2014] [Indexed: 12/23/2022]
Abstract
Mechanotransduction describes how a cell senses and interacts with its environment. The concept originated in adhesion biology where adhesion receptors, integrins, facilitate force transmission between the extracellular matrix and the intracellular actin cytoskeleton. Indeed, during any adhesive contacts, cells do exert mechanical force. Hence, the probing of the local environment by cells results in mechanical cues that contribute to cellular functions and cell fate decisions such as migration, proliferation, differentiation and apoptosis. On the molecular level, mechanical forces can rearrange proteins laterally within the membrane, regulate their activity by inducing conformational changes and probe the mechanical properties and bond strength of receptor-ligands. From this point of view, it appears surprising that molecular forces have been largely overlooked in membrane organisation and ligand discrimination processes in lymphocytes. During T cell activation, the T cell receptor recognises and distinguishes antigenic from benign endogenous peptides to initiate the reorganisation of membrane proteins into signalling clusters within the immunological synapse. In this review, we asked whether characteristics of fibroblast force sensing could be applied to immune cell antigen recognition and signalling, and outline state-of-the-art experimental strategies for studying forces in the context of membrane organisation. This article is part of a Special Issue entitled: Nanoscale membrane orgainisation and signalling.
Collapse
Affiliation(s)
- Enrico Klotzsch
- Centre for Vascular Research, ARC Centre of Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia.
| | - Johannes Stiegler
- Centre for Vascular Research, ARC Centre of Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia
| | - Eldad Ben-Ishay
- Centre for Vascular Research, ARC Centre of Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia
| | - Katharina Gaus
- Centre for Vascular Research, ARC Centre of Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|