101
|
Jenkins E, Körbel M, O'Brien-Ball C, McColl J, Chen KY, Kotowski M, Humphrey J, Lippert AH, Brouwer H, Santos AM, Lee SF, Davis SJ, Klenerman D. Antigen discrimination by T cells relies on size-constrained microvillar contact. Nat Commun 2023; 14:1611. [PMID: 36959206 PMCID: PMC10036606 DOI: 10.1038/s41467-023-36855-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 02/21/2023] [Indexed: 03/25/2023] Open
Abstract
T cells use finger-like protrusions called 'microvilli' to interrogate their targets, but why they do so is unknown. To form contacts, T cells must overcome the highly charged, barrier-like layer of large molecules forming a target cell's glycocalyx. Here, T cells are observed to use microvilli to breach a model glycocalyx barrier, forming numerous small (<0.5 μm diameter) contacts each of which is stabilized by the small adhesive protein CD2 expressed by the T cell, and excludes large proteins including CD45, allowing sensitive, antigen dependent TCR signaling. In the absence of the glycocalyx or when microvillar contact-size is increased by enhancing CD2 expression, strong signaling occurs that is no longer antigen dependent. Our observations suggest that, modulated by the opposing effects of the target cell glycocalyx and small adhesive proteins, the use of microvilli equips T cells with the ability to effect discriminatory receptor signaling.
Collapse
Affiliation(s)
- Edward Jenkins
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Markus Körbel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Caitlin O'Brien-Ball
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - James McColl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Kevin Y Chen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Mateusz Kotowski
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Jane Humphrey
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Anna H Lippert
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Heather Brouwer
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Steven F Lee
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
102
|
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
|
103
|
Anto NP, Muraleedharan A, Nath PR, Sun Z, Keasar C, Livneh E, Braiman A, Altman A, Kong KF, Isakov N. The Peptidyl-Prolyl cis-trans isomerase, Pin1, associates with Protein Kinase C θ via a critical Phospho-Thr-Pro motif in the V3 regulatory domain. Front Immunol 2023; 14:1126464. [PMID: 36969236 PMCID: PMC10031136 DOI: 10.3389/fimmu.2023.1126464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Protein kinase C-θ (PKCθ) is a member of the novel PKC subfamily known for its selective and predominant expression in T lymphocytes where it regulates essential functions required for T cell activation and proliferation. Our previous studies provided a mechanistic explanation for the recruitment of PKCθ to the center of the immunological synapse (IS) by demonstrating that a proline-rich (PR) motif within the V3 region in the regulatory domain of PKCθ is necessary and sufficient for PKCθ IS localization and function. Herein, we highlight the importance of Thr335-Pro residue in the PR motif, the phosphorylation of which is key in the activation of PKCθ and its subsequent IS localization. We demonstrate that the phospho-Thr335-Pro motif serves as a putative binding site for the peptidyl-prolyl cis-trans isomerase (PPIase), Pin1, an enzyme that specifically recognizes peptide bonds at phospho-Ser/Thr-Pro motifs. Binding assays revealed that mutagenesis of PKCθ-Thr335-to-Ala abolished the ability of PKCθ to interact with Pin1, while Thr335 replacement by a Glu phosphomimetic, restored PKCθ binding to Pin1, suggesting that Pin1-PKCθ association is contingent upon the phosphorylation of the PKCθ-Thr335-Pro motif. Similarly, the Pin1 mutant, R17A, failed to associate with PKCθ, suggesting that the integrity of the Pin1 N-terminal WW domain is a requisite for Pin1-PKCθ interaction. In silico docking studies underpinned the role of critical residues in the Pin1-WW domain and the PKCθ phospho-Thr335-Pro motif, to form a stable interaction between Pin1 and PKCθ. Furthermore, TCR crosslinking in human Jurkat T cells and C57BL/6J mouse-derived splenic T cells promoted a rapid and transient formation of Pin1-PKCθ complexes, which followed a T cell activation-dependent temporal kinetic, suggesting a role for Pin1 in PKCθ-dependent early activation events in TCR-triggered T cells. PPIases that belong to other subfamilies, i.e., cyclophilin A or FK506-binding protein, failed to associate with PKCθ, indicating the specificity of the Pin1-PKCθ association. Fluorescent cell staining and imaging analyses demonstrated that TCR/CD3 triggering promotes the colocalization of PKCθ and Pin1 at the cell membrane. Furthermore, interaction of influenza hemagglutinin peptide (HA307-319)-specific T cells with antigen-fed antigen presenting cells (APCs) led to colocalization of PKCθ and Pin1 at the center of the IS. Together, we point to an uncovered function for the Thr335-Pro motif within the PKCθ-V3 regulatory domain to serve as a priming site for its activation upon phosphorylation and highlight its tenability to serve as a regulatory site for the Pin1 cis-trans isomerase.
Collapse
Affiliation(s)
- Nikhil Ponnoor Anto
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Amitha Muraleedharan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Pulak Ranjan Nath
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Zuoming Sun
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chen Keasar
- The Department of Computer Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Etta Livneh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Immunology, San Diego, CA, United States
| | - Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Immunology, San Diego, CA, United States
| | - Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- *Correspondence: Noah Isakov,
| |
Collapse
|
104
|
Beppler C, Eichorst J, Marchuk K, Cai E, Castellanos CA, Sriram V, Roybal KT, Krummel MF. Hyperstabilization of T cell microvilli contacts by chimeric antigen receptors. J Cell Biol 2023; 222:e202205118. [PMID: 36520493 PMCID: PMC9757849 DOI: 10.1083/jcb.202205118] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/25/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
T cells typically recognize their ligands using a defined cell biology-the scanning of their membrane microvilli (MV) to palpate their environment-while that same membrane scaffolds T cell receptors (TCRs) that can signal upon ligand binding. Chimeric antigen receptors (CARs) present both a therapeutic promise and a tractable means to study the interplay between receptor affinity, MV dynamics and T cell function. CARs are often built using single-chain variable fragments (scFvs) with far greater affinity than that of natural TCRs. We used high-resolution lattice lightsheet (LLS) and total internal reflection fluorescence (TIRF) imaging to visualize MV scanning in the context of variations in CAR design. This demonstrated that conventional CARs hyper-stabilized microvillar contacts relative to TCRs. Reducing receptor affinity, antigen density, and/or multiplicity of receptor binding sites normalized microvillar dynamics and synapse resolution, and effector functions improved with reduced affinity and/or antigen density, highlighting the importance of understanding the underlying cell biology when designing receptors for optimal antigen engagement.
Collapse
Affiliation(s)
- Casey Beppler
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| | - John Eichorst
- Biological Imaging Development CoLab, University of California, San Francisco, San Francisco, CA, USA
| | - Kyle Marchuk
- Biological Imaging Development CoLab, University of California, San Francisco, San Francisco, CA, USA
| | - En Cai
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos A. Castellanos
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | | | - Kole T. Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Matthew F. Krummel
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
105
|
White D, Cote-Martin A, Bleck M, Garaffa N, Shaaban A, Wu H, Liu D, Young D, Scheer J, Lorenz IC, Nixon A, Fine JS, Byrne FR, Mbow ML, Moreno-Garcia ME. Programmed Cell Death-1 (PD-1) anchoring to the GPI-linked co-receptor CD48 reveals a novel mechanism to modulate PD-1-dependent inhibition of human T cells. Mol Immunol 2023; 156:31-38. [PMID: 36889184 DOI: 10.1016/j.molimm.2023.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023]
Abstract
Activation of PD-1 by anchoring it to Antigen Receptor (AR) components or associated co-receptors represents an attractive approach to treat autoimmune conditions. In this study, we provide evidence that CD48, a common lipid raft and Src kinase-associated coreceptor, induces significant Src kinase-dependent activation of PD-1 upon crosslinking, while CD71, a receptor excluded from these compartments, does not. Functionally, using bead-conjugated antibodies we demonstrate that CD48-dependent activation of PD-1 inhibits proliferation of AR-induced primary human T cells, and similarly, PD-1 activation using PD-1/CD48 bispecific antibodies inhibits IL-2, enhances IL-10 secretion, and reduces NFAT activation in primary human and Jurkat T cells, respectively. As a whole, CD48-dependent activation of PD-1 represents a novel mechanism to fine tune T cell activation, and by functionally anchoring PD-1 with receptors other than AR, this study provides a conceptual framework for rational development of novel therapies that activate inhibitory checkpoint receptors for treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Della White
- Departments of Immunology and Respiratory Research, USA.
| | | | - Marina Bleck
- Departments of Immunology and Respiratory Research, USA
| | | | | | - Helen Wu
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Dongmei Liu
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - David Young
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Justin Scheer
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Ivo C Lorenz
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Andrew Nixon
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Jay S Fine
- Departments of Immunology and Respiratory Research, USA
| | | | - M Lamine Mbow
- Departments of Immunology and Respiratory Research, USA; Cancer Immunology and Immune Modulation, USA
| | | |
Collapse
|
106
|
Chien CY, Lin JC, Huang CY, Hsu CY, Yang KC, Chattopadhyay S, Nikoloutsos N, Hsieh PCH, Hu CMJ. In Situ Hydrogelation of Cellular Monolayers Enables Conformal Biomembrane Functionalization for Xeno-Free Feeder Substrate Engineering. Adv Healthc Mater 2023; 12:e2201708. [PMID: 36455286 DOI: 10.1002/adhm.202201708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/14/2022] [Indexed: 12/03/2022]
Abstract
The intricate functionalities of cellular membranes have inspired strategies for deriving and anchoring cell-surface components onto solid substrates for biological studies, biosensor applications, and tissue engineering. However, introducing conformal and right-side-out cell membrane coverage onto planar substrates requires cumbersome protocols susceptible to significant device-to-device variability. Here, a facile approach for biomembrane functionalization of planar substrates is demonstrated by subjecting confluent cellular monolayer to intracellular hydrogel polymerization. The resulting cell-gel hybrid, herein termed GELL (gelated cell), exhibits extraordinary stability and retains the structural integrity, membrane fluidity, membrane protein mobility, and topology of living cells. In assessing the utility of GELL layers as a tissue engineering feeder substrate for stem cell maintenance, GELL feeder prepared from primary mouse embryonic fibroblasts not only preserves the stemness of murine stem cells but also exhibits advantages over live feeder cells owing to the GELL's inanimate, non-metabolizing nature. The preparation of a xeno-free feeder substrate devoid of non-human components is further shown with HeLa cells, and the resulting HeLa GELL feeder effectively sustains the growth and stemness of both murine and human induced pluripotent stem cells. The study highlights a novel bio-functionalization strategy that introduces new opportunities for tissue engineering and other biomedical applications.
Collapse
Affiliation(s)
- Chen-Ying Chien
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Jung-Chen Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Ching-Ying Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Chung-Yao Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | - Kai-Chieh Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | - Saborni Chattopadhyay
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | | | | | - Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
107
|
Chew PY, Joseph JA, Collepardo-Guevara R, Reinhardt A. Thermodynamic origins of two-component multiphase condensates of proteins. Chem Sci 2023; 14:1820-1836. [PMID: 36819870 PMCID: PMC9931050 DOI: 10.1039/d2sc05873a] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023] Open
Abstract
Intracellular condensates are highly multi-component systems in which complex phase behaviour can ensue, including the formation of architectures comprising multiple immiscible condensed phases. Relying solely on physical intuition to manipulate such condensates is difficult because of the complexity of their composition, and systematically learning the underlying rules experimentally would be extremely costly. We address this challenge by developing a computational approach to design pairs of protein sequences that result in well-separated multilayered condensates and elucidate the molecular origins of these compartments. Our method couples a genetic algorithm to a residue-resolution coarse-grained protein model. We demonstrate that we can design protein partners to form multiphase condensates containing naturally occurring proteins, such as the low-complexity domain of hnRNPA1 and its mutants, and show how homo- and heterotypic interactions must differ between proteins to result in multiphasicity. We also show that in some cases the specific pattern of amino-acid residues plays an important role. Our findings have wide-ranging implications for understanding and controlling the organisation, functions and material properties of biomolecular condensates.
Collapse
Affiliation(s)
- Pin Yu Chew
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| | - Jerelle A Joseph
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
- Department of Physics, University of Cambridge Cambridge CB3 0HE UK
- Department of Genetics, University of Cambridge Cambridge CB2 3EH UK
| | - Rosana Collepardo-Guevara
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
- Department of Physics, University of Cambridge Cambridge CB3 0HE UK
- Department of Genetics, University of Cambridge Cambridge CB2 3EH UK
| | - Aleks Reinhardt
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| |
Collapse
|
108
|
Li K, Chen Y, Zhu N, Chen S, Jia M, Xue L, Hao M, Zhang C. Real-time detection of T cell activation by visualizing TCR nanoclusters with a cholesterol derived aggregation-induced emission probe. Eur J Med Chem 2023; 247:115073. [PMID: 36603511 DOI: 10.1016/j.ejmech.2022.115073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/18/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022]
Abstract
Successful T-cell based immunotherapy usually depends on the activation of T cells. Most of commonly used methods for assessing T cell activity rely on the antibody-based technology, which focus on detecting protein-centered activation markers, including CD25, cytokines and so on. However, these methods always involve tedious sample-preparation process, labor-consuming and costly, which could not be utilized in real-time detection. The T cell receptor (TCR) clustering is another kind of essential T cell activation marker on the membrane, which increases during the activation state of T cells. We herein developed a cholesterol derived aggregation-induced emission (AIE) fluorescent probe (R-TPE-PEG-Chol) for detecting T cell activation in real-time. Five probes were first designed and synthesized and among them COOH-TPE-PEG-Chol displayed the best imaging effects, which had no significant impact on the key physiological functions of T cells. In addition, we have proved that COOH-TPE-PEG-Chol was introduced onto the naïve T cell membrane in its molecularly dissolved form without fluorescent emission. While during T cell activation, the formation of TCR nanoclusters would induce aggregation of membrane cholesterol, which could provoke the fluorescence signal of the COOH-TPE-PEG-Chol due to the AIE characteristic. Moreover, the enhancement of the fluorescence intensity was positively related to the activation state of T cells. Our study demonstrated the concept of cholesterol-derived AIE fluorescent probes for deciphering the spatiotemporal arrangements of TCR on the membrane during T cell activation, and consequently provided a novel and complementary strategy for detecting T cell activation in real-time.
Collapse
Affiliation(s)
- Kaiming Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| | - Yue Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Nianci Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| | - Sijia Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| | - Meng Jia
- Nanjing University, School of Life Sciences, Nanjing, 210093, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| | - Meixi Hao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China.
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China.
| |
Collapse
|
109
|
Kim HR, Park JS, Soh WC, Kim NY, Moon HY, Lee JS, Jun CD. T Cell Microvilli: Finger-Shaped External Structures Linked to the Fate of T Cells. Immune Netw 2023; 23:e3. [PMID: 36911802 PMCID: PMC9995986 DOI: 10.4110/in.2023.23.e3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/11/2023] [Accepted: 02/11/2023] [Indexed: 03/07/2023] Open
Abstract
Microvilli are outer membrane organelles that contain cross-linked filamentous actin. Unlike well-characterized epithelial microvilli, T-cell microvilli are dynamic similar to those of filopodia, which grow and shrink intermittently via the alternate actin-assembly and -disassembly. T-cell microvilli are specialized for sensing Ags on the surface of Ag-presenting cells (APCs). Thus, these finger-shaped microprotrusions contain many signaling-related proteins and can serve as a signaling platforms that induce intracellular signals. However, they are not limited to sensing external information but can provide sites for parts of the cell-body to tear away from the cell. Cells are known to produce many types of extracellular vesicles (EVs), such as exosomes, microvesicles, and membrane particles. T cells also produce EVs, but little is known about under what conditions T cells generate EVs and which types of EVs are released. We discovered that T cells produce few exosomes but release large amounsts of microvilli-derived particles during physical interaction with APCs. Although much is unanswered as to why T cells use the same organelles to sense Ags or to produce EVs, these events can significantly affect T cell fate, including clonal expansion and death. Since TCRs are localized at microvilli tips, this membrane event also raises a new question regarding long-standing paradigm in T cell biology; i.e., surface TCR downmodulation following T cell activation. Since T-cell microvilli particles carry T-cell message to their cognate partner, these particles are termed T-cell immunological synaptosomes (TISs). We discuss the potential physiological role of TISs and their application to immunotherapies.
Collapse
Affiliation(s)
- Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Division of Rare and Refractory Cancer, Tumor Immunology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Jeong-Su Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Won-Chang Soh
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Na-Young Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Hyun-Yoong Moon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Ji-Su Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| |
Collapse
|
110
|
Espie D, Donnadieu E. CAR T-cell behavior and function revealed by real-time imaging. Semin Immunopathol 2023; 45:229-239. [PMID: 36688965 DOI: 10.1007/s00281-023-00983-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023]
Abstract
Adoptive transfer of T-cells expressing chimeric antigen receptors (CAR) has shown remarkable clinical efficacy against advanced B-cell malignancies. Nonetheless, the field of CAR T-cells is currently facing several major challenges. In particular, the CAR T-cell strategy has not yet produced favorable clinical responses when targeting solid tumors. In this context, it is of paramount importance to understand the determinants that limit the efficacy of T-cell-based immunotherapy. Characterization of CAR T-cells is usually based on flow cytometry and whole-transcriptome profiling. These approaches have been very valuable to determine intrinsic elements that condition T-cell ability to proliferate and expand. However, they do not take into account spatial and kinetic aspects of T-cell responses. In particular, in order to control tumor growth, CAR T-cells need to enter into the tumor, migrate within a complex tumor environment, and form productive conjugates with their targets. Advanced imaging techniques combined with innovative preclinical models represent promising tools to uncover the dynamics of CAR T-cells. In this review, we will discuss recent results on the biology of engineered T-cells that have been obtained with real-time imaging microscopy. Important notions have emerged from these imaging-based studies, such as the multi-killing potential of CAR T-cells. Finally, we will highlight how imaging techniques combined with other tools can solve remaining unresolved questions in the field of engineered T-cells.
Collapse
Affiliation(s)
- David Espie
- Université Paris Cité, CNRS, INSERM, Equipe Labellisée Ligue Contre le Cancer, Institut Cochin, INSERM U1016, 22 rue Méchain, F-75014, Paris, France.,Invectys, Paris, France
| | - Emmanuel Donnadieu
- Université Paris Cité, CNRS, INSERM, Equipe Labellisée Ligue Contre le Cancer, Institut Cochin, INSERM U1016, 22 rue Méchain, F-75014, Paris, France.
| |
Collapse
|
111
|
Cerny O. Quantification of interaction frequency between antigen-presenting cells and T cells by conjugation assay. Methods Cell Biol 2023; 173:65-75. [PMID: 36653086 DOI: 10.1016/bs.mcb.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Interaction between an antigen-presenting cell and a T cell, and their subsequent conjugation are a prerequisite for the formation of the immunological synapse and productive, antigen-dependent activation of T cells. This initial interaction is accompanied by recognition of the presented antigen by the T cell receptor, and by changes in the morphology of the interacting cells and in actin cytoskeleton structure in the site of interaction. The experimental protocol below describes a simple assay for quantitative assessment of antigen-presenting cells-T cell conjugation using confocal microscopy or flow cytometry.
Collapse
Affiliation(s)
- Ondrej Cerny
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
112
|
Gan J, Cho JH, Lee R, Naghizadeh A, Poon LY, Wang E, Hui Z, Liu D. Methods of Machine Learning-Based Chimeric Antigen Receptor Immunological Synapse Quality Quantification. Methods Mol Biol 2023; 2654:493-502. [PMID: 37106203 PMCID: PMC10812845 DOI: 10.1007/978-1-0716-3135-5_32] [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] [Indexed: 04/29/2023]
Abstract
Chimeric Antigen Receptor (CAR)-mediated immunotherapy shows promising results for refractory blood cancers. Currently, six CAR-T drugs have been approved by U.S. Food and Drug Administration (FDA). Theoretically, CAR-T cells must form an effective immunological synapse (IS, an interface between effective cells and their target cells) with their susceptible tumor cells to eliminate tumor cells. Previous studies show that CAR IS quality can be used as a predictive functional biomarker for CAR-T immunotherapies. However, quantification of CAR-T IS quality is clinically challenging. Machine learning (ML)-based CAR-T IS quality quantification has been proposed previously.Here, we show an easy-to-use, step-by-step approach to predicting the efficacy of CAR-modified cells using ML-based CAR IS quality quantification. This approach will guide the users on how to use ML-based CAR IS quality quantification in detail, which include: how to image CAR IS on the glass-supported planar lipid bilayer, how to define the CAR IS focal plane, how to segment the CAR IS images, and how to quantify the IS quality using ML-based algorithms.This approach will significantly enhance the accuracy and proficiency of CAR IS prediction in research.
Collapse
Affiliation(s)
- Julian Gan
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Jong Hyun Cho
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Newark, NJ, USA
| | - Ryan Lee
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Alireza Naghizadeh
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Ling Yue Poon
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Ethan Wang
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Zachary Hui
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA.
- Center for Immunity and Inflammation, New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
113
|
Kim HR, Park JS, Kim NY, Jun CD. T Cell Immunological Synaptosomes: Definition and Isolation. Methods Mol Biol 2023; 2654:201-215. [PMID: 37106184 DOI: 10.1007/978-1-0716-3135-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
In addition to microvilli's role as structural scaffold for TCR clustering, we recently discovered a novel function as message senders. We found that microvilli are separated from the T cell body shortly upon TCR stimulation and vesiculated to form T cell microvilli particles (TMPs), a new type of membrane vesicles. TMPs and synaptic ectosomes, which bud from the synaptic cleft, constitute "T cell immunological synaptosomes (TISs)" and act as conveyors of T cell messages or traits to cognate antigen-presenting cells. In practice, it is almost impossible to distinguish between TMPs and synaptic ectosomes. Here, we describe a newly developed protocol to isolate TISs from activated T cells using antibody-immobilized agarose beads and density gradient ultracentrifugation. We further describe the methods for TIS quantification with flow cytometry and to evaluate TIS efficacy on dendritic cells.
Collapse
Affiliation(s)
- Hye-Ran Kim
- School of Life Sciences, Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
- Division of Rare and Refractory Cancer, Immuno-oncology, Research Institute, National Cancer Center, Goyang, South Korea
| | - Jeong-Su Park
- School of Life Sciences, Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Na-Young Kim
- School of Life Sciences, Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Chang-Duk Jun
- School of Life Sciences, Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea.
| |
Collapse
|
114
|
Abdelrahman A, Smith AS, Sengupta K. Observing Membrane and Cell Adhesion via Reflection Interference Contrast Microscopy. Methods Mol Biol 2023; 2654:123-135. [PMID: 37106179 DOI: 10.1007/978-1-0716-3135-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Reflection interference contrast microscopy (RICM) is an optical microscopy technique ideally suited for imaging adhesion. While RICM (and the closely related interference reflection microscopy (IRM)) has been extensively used qualitatively or semiquantitatively to image cells, including immune cells, it can also be used quantitatively to measure membrane to surface distance, especially for model membranes. Here, we present a protocol for RICM and IRM imaging and the details of semiquantitative and quantitative analysis.
Collapse
Affiliation(s)
- Ahmed Abdelrahman
- Aix Marseille University, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | - Ana-Sunčana Smith
- PULS Group, Department of Physics, Centre for Computational Materials and Processes, Friedrich Alexander University Erlangen-Nürnberg, IZNF, Erlangen, Germany.
- Group for Computational Life Sciences, Division of Physical Chemistry, Ruđer Bošković Institute, Zagreb, Croatia.
| | - Kheya Sengupta
- Aix Marseille University, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| |
Collapse
|
115
|
Mørch AM, Schneider F. Investigating Diffusion Dynamics and Interactions with Scanning Fluorescence Correlation Spectroscopy (sFCS). Methods Mol Biol 2023; 2654:61-89. [PMID: 37106176 DOI: 10.1007/978-1-0716-3135-5_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Activation of immune cells and formation of immunological synapses (IS) rely critically on the reorganization of the plasma membrane. These highly orchestrated processes are driven by diffusion and oligomerization dynamics, as well as by single molecule interactions. While slow macro- and meso-scale changes in organization can be observed with conventional imaging, fast nano-scale dynamics are often missed with traditional approaches, but resolving them is, nonetheless, essential to understand the underlying biological mechanisms at play. Here, we describe the use of scanning fluorescence correlation spectroscopy (sFCS) and scanning fluorescence cross-correlation spectroscopy (sFCCS) to study reorganization and changes in molecular diffusion dynamics and interactions during IS formation and in other biological settings. We focus on the practical aspects of the measurements including calibration and alignment of the optical setup, present a comprehensive protocol to perform the measurements, and provide data analysis pipelines and strategies. Finally, we show an exemplary application of the technology to studying Lck diffusion during T-cell signaling.
Collapse
Affiliation(s)
| | - Falk Schneider
- Translational Imaging Center, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
116
|
Lanna A, D'Ambra C. Detection of Telomere Transfer at Immunological Synapse. Methods Mol Biol 2023; 2654:251-261. [PMID: 37106187 DOI: 10.1007/978-1-0716-3135-5_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Eukaryotes solve the DNA-end replication problem synthesizing hexameric chromosome ends known as telomeres. Recent studies have uncovered unexpected functions of telomeres in linking synaptic signaling and vesicle transport, with at least one pathway directly involved in transferring telomeres through the immune synapse. These emerging forms of cellular communication may originate a new class of antiaging interventions based on telomere transplants.
Collapse
|
117
|
Alatoom A, ElGindi M, Sapudom J, Teo JCM. The T Cell Journey: A Tour de Force. Adv Biol (Weinh) 2023; 7:e2200173. [PMID: 36190140 DOI: 10.1002/adbi.202200173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/30/2022] [Indexed: 11/07/2022]
Abstract
T cells act as the puppeteers in the adaptive immune response, and their dysfunction leads to the initiation and progression of pathological conditions. During their lifetime, T cells experience myriad forces that modulate their effector functions. These forces are imposed by interacting cells, surrounding tissues, and shear forces from fluid movement. In this review, a journey with T cells is made, from their development to their unique characteristics, including the early studies that uncovered their mechanosensitivity. Then the studies pertaining to the responses of T cell activation to changes in antigen-presenting cells' physical properties, to their immediate surrounding extracellular matrix microenvironment, and flow conditions are highlighted. In addition, it is explored how pathological conditions like the tumor microenvironment can hinder T cells and allow cancer cells to escape elimination.
Collapse
Affiliation(s)
- Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE.,Department of Mechanical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE
| | - Jeremy C M Teo
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE.,Department of Mechanical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.,Department of Biomedical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| |
Collapse
|
118
|
Abstract
Here, we describe a method, which we term "shadow imaging," to analyze the secretions of individual cells at immune synapses or other cell contacts. Following immune synapse formation and cellular activation on ligand-rich slides, the position of each cell is recorded using a pulsed immunofluorescence stain against the proteins on the ligand-rich slide surface. The pulsed stain does not penetrate the synaptic cleft, resulting in an unlabeled region or "shadow" beneath cells that is retained following cellular detachment. The secreted components, such as perforin, exosomes, or other types of extracellular vesicles, are retained on the slide and can be analyzed on a single-cell basis using immunofluorescence. The ability to identify single cells secreting different combinations of particles, proteins, and vesicles enables us to better understand the heterogeneity in immune cell secretions and can be used as a novel approach for phenotyping cell populations.
Collapse
Affiliation(s)
- Ashley R Ambrose
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Khodor S Hazime
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Department of Life Sciences, Imperial College London, London, UK
| | - Daniel M Davis
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
- Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
119
|
Bispecific Antibody Format and the Organization of Immunological Synapses in T Cell-Redirecting Strategies for Cancer Immunotherapy. Pharmaceutics 2022; 15:pharmaceutics15010132. [PMID: 36678761 PMCID: PMC9863865 DOI: 10.3390/pharmaceutics15010132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023] Open
Abstract
T cell-redirecting strategies have emerged as effective cancer immunotherapy approaches. Bispecific antibodies (bsAbs) are designed to specifically recruit T cells to the tumor microenvironment and induce the assembly of the immunological synapse (IS) between T cells and cancer cells or antigen-presenting cells. The way that the quality of the IS might predict the effectiveness of T cell-redirecting strategies, including those mediated by bsAbs or by chimeric antigen receptors (CAR)-T cells, is currently under discussion. Here we review the organization of the canonical IS assembled during natural antigenic stimulation through the T cell receptor (TCR) and to what extent different bsAbs induce T cell activation, canonical IS organization, and effector function. Then, we discuss how the biochemical parameters of different formats of bsAbs affect the effectivity of generating an antigen-induced canonical IS. Finally, the quality of the IS assembled by bsAbs and monoclonal antibodies or CAR-T cells are compared, and strategies to improve bsAb-mediated T cell-redirecting strategies are discussed.
Collapse
|
120
|
McAffee DB, O'Dair MK, Lin JJ, Low-Nam ST, Wilhelm KB, Kim S, Morita S, Groves JT. Discrete LAT condensates encode antigen information from single pMHC:TCR binding events. Nat Commun 2022; 13:7446. [PMID: 36460640 PMCID: PMC9718779 DOI: 10.1038/s41467-022-35093-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
LAT assembly into a two-dimensional protein condensate is a prominent feature of antigen discrimination by T cells. Here, we use single-molecule imaging techniques to resolve the spatial position and temporal duration of each pMHC:TCR molecular binding event while simultaneously monitoring LAT condensation at the membrane. An individual binding event is sufficient to trigger a LAT condensate, which is self-limiting, and neither its size nor lifetime is correlated with the duration of the originating pMHC:TCR binding event. Only the probability of the LAT condensate forming is related to the pMHC:TCR binding dwell time. LAT condenses abruptly, but after an extended delay from the originating binding event. A LAT mutation that facilitates phosphorylation at the PLC-γ1 recruitment site shortens the delay time to LAT condensation and alters T cell antigen specificity. These results identify a function for the LAT protein condensation phase transition in setting antigen discrimination thresholds in T cells.
Collapse
Affiliation(s)
- Darren B McAffee
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Mark K O'Dair
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jenny J Lin
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Shalini T Low-Nam
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Kiera B Wilhelm
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Sungi Kim
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Shumpei Morita
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jay T Groves
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA.
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore.
| |
Collapse
|
121
|
Teppert K, Wang X, Anders K, Evaristo C, Lock D, Künkele A. Joining Forces for Cancer Treatment: From "TCR versus CAR" to "TCR and CAR". Int J Mol Sci 2022; 23:14563. [PMID: 36498890 PMCID: PMC9739809 DOI: 10.3390/ijms232314563] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
T cell-based immunotherapy has demonstrated great therapeutic potential in recent decades, on the one hand, by using tumor-infiltrating lymphocytes (TILs) and, on the other hand, by engineering T cells to obtain anti-tumor specificities through the introduction of either engineered T cell receptors (TCRs) or chimeric antigen receptors (CARs). Given the distinct design of both receptors and the type of antigen that is encountered, the requirements for proper antigen engagement and downstream signal transduction by TCRs and CARs differ. Synapse formation and signal transduction of CAR T cells, despite further refinement of CAR T cell designs, still do not fully recapitulate that of TCR T cells and might limit CAR T cell persistence and functionality. Thus, deep knowledge about the molecular differences in CAR and TCR T cell signaling would greatly advance the further optimization of CAR designs and elucidate under which circumstances a combination of both receptors would improve the functionality of T cells for cancer treatment. Herein, we provide a comprehensive review about similarities and differences by directly comparing the architecture, synapse formation and signaling of TCRs and CARs, highlighting the knowns and unknowns. In the second part of the review, we discuss the current status of combining CAR and TCR technologies, encouraging a change in perspective from "TCR versus CAR" to "TCR and CAR".
Collapse
Affiliation(s)
- Karin Teppert
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Xueting Wang
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Kathleen Anders
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - César Evaristo
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Dominik Lock
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Annette Künkele
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| |
Collapse
|
122
|
Wen Y, Ma J. Phase separation drives the formation of biomolecular condensates in the immune system. Front Immunol 2022; 13:986589. [PMID: 36439121 PMCID: PMC9685520 DOI: 10.3389/fimmu.2022.986589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/19/2022] [Indexed: 08/12/2023] Open
Abstract
When the external conditions change, such as the temperature or the pressure, the multi-component system sometimes separates into several phases with different components and structures, which is called phase separation. Increasing studies have shown that cells condense related biomolecules into independent compartments in order to carry out orderly and efficient biological reactions with the help of phase separation. Biomolecular condensates formed by phase separation play a significant role in a variety of cellular processes, including the control of signal transduction, the regulation of gene expression, and the stress response. In recent years, many phase separation events have been discovered in the immune response process. In this review, we provided a comprehensive and detailed overview of the role and mechanism of phase separation in the innate and adaptive immune responses, which will help the readers to appreciate the advance and importance of this field.
Collapse
Affiliation(s)
- Yuqing Wen
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Jian Ma
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| |
Collapse
|
123
|
Behrens LM, van Egmond M, van den Berg TK. Neutrophils as immune effector cells in antibody therapy in cancer. Immunol Rev 2022; 314:280-301. [PMID: 36331258 DOI: 10.1111/imr.13159] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Tumor-targeting monoclonal antibodies are available for a number of cancer cell types (over)expressing the corresponding tumor antigens. Such antibodies can limit tumor progression by different mechanisms, including direct growth inhibition and immune-mediated mechanisms, in particular complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis, and antibody-dependent cellular cytotoxicity (ADCC). ADCC can be mediated by various types of immune cells, including neutrophils, the most abundant leukocyte in circulation. Neutrophils express a number of Fc receptors, including Fcγ- and Fcα-receptors, and can therefore kill tumor cells opsonized with either IgG or IgA antibodies. In recent years, important insights have been obtained with respect to the mechanism(s) by which neutrophils engage and kill antibody-opsonized cancer cells and these findings are reviewed here. In addition, we consider a number of additional ways in which neutrophils may affect cancer progression, in particular by regulating adaptive anti-cancer immunity.
Collapse
Affiliation(s)
- Leonie M. Behrens
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Vrije Universiteit Amsterdam HV Amsterdam The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology HV Amsterdam The Netherlands
- Amsterdam institute for Infection and Immunity, Cancer Immunology HV Amsterdam The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Vrije Universiteit Amsterdam HV Amsterdam The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology HV Amsterdam The Netherlands
- Amsterdam institute for Infection and Immunity, Cancer Immunology HV Amsterdam The Netherlands
- Department of Surgery, Amsterdam UMC Vrije Universiteit Amsterdam HV Amsterdam The Netherlands
| | | |
Collapse
|
124
|
Duwa R, Pokhrel RH, Banstola A, Pandit M, Shrestha P, Jeong JH, Chang JH, Yook S. T-cell engaging poly(lactic-co-glycolic acid) nanoparticles as a modular platform to induce a potent cytotoxic immunogenic response against PD-L1 overexpressing cancer. Biomaterials 2022; 291:121911. [DOI: 10.1016/j.biomaterials.2022.121911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022]
|
125
|
Anikeeva N, Steblyanko M, Kuri-Cervantes L, Buggert M, Betts MR, Sykulev Y. The immune synapses reveal aberrant functions of CD8 T cells during chronic HIV infection. Nat Commun 2022; 13:6436. [PMID: 36307445 PMCID: PMC9616955 DOI: 10.1038/s41467-022-34157-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 10/14/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic HIV infection causes persistent low-grade inflammation that induces premature aging of the immune system including senescence of memory and effector CD8 T cells. To uncover the reasons of gradually diminished potency of CD8 T cells from people living with HIV, here we expose the T cells to planar lipid bilayers containing ligands for T-cell receptor and a T-cell integrins and analyze the cellular morphology, dynamics of synaptic interface formation and patterns of the cellular degranulation. We find a large fraction of phenotypically naive T cells from chronically infected people are capable to form mature synapse with focused degranulation, a signature of a differentiated T cells. Further, differentiation of aberrant naive T cells may lead to the development of anomalous effector T cells undermining their capacity to control HIV and other pathogens that could be contained otherwise.
Collapse
Affiliation(s)
- Nadia Anikeeva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria Steblyanko
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcus Buggert
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Michael R Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuri Sykulev
- Departments of Immunology and Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sydney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
126
|
Orlik C, Berschneider KM, Jahraus B, Niesler B, Balta E, Schäkel K, Schröder-Braunstein J, Souto-Carneiro MM, Samstag Y. Keratinocyte-induced costimulation of human T cells through CD6 - but not CD2 - activates mTOR and prevents oxidative stress. Front Immunol 2022; 13:1016112. [PMID: 36353616 PMCID: PMC9639098 DOI: 10.3389/fimmu.2022.1016112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/27/2022] [Indexed: 11/26/2022] Open
Abstract
In psoriasis and other inflammatory skin diseases, keratinocytes (KCs) secrete chemokines that attract T cells, which, in turn, cause epidermal hyperplasia by secreting proinflammatory cytokines. To date, it remains unclear whether skin-homing T cells, particularly memory T cells, can also be activated by direct cell contact with KCs. In this study, we demonstrated the ability of primary human KCs to activate human memory T cells directly by transmitting costimulatory signals through the CD6/CD166/CD318 axis. Interestingly, despite being negative for CD80/CD86, KCs initiate a metabolic shift within T cells. Blockade of the CD6/CD166/CD318 axis prevents mammalian target of rapamycin activation and T cell proliferation but promotes oxidative stress and aerobic glycolysis. In addition, it diminishes formation of central memory T cells. Importantly, although KC-mediated costimulation by CD2/CD58 also activates T cells, it cannot compensate for the lack of CD6 costimulation. Therefore, KCs likely differentially regulate T cell functions in the skin through two distinct costimulatory receptors: CD6 and CD2. This may at least in part explain the divergent effects observed when treating inflammatory skin diseases with antibodies to CD6 versus CD2. Moreover, our findings may provide a molecular basis for selective interference with either CD6/CD166/CD318, or CD2/CD58, or both to specifically treat different types of inflammatory skin diseases.
Collapse
Affiliation(s)
- Christian Orlik
- Institute of Immunology, Section Molecular Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karina M. Berschneider
- Institute of Immunology, Section Molecular Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Beate Jahraus
- Institute of Immunology, Section Molecular Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Beate Niesler
- Institute of Human Genetics, Department of Human Molecular Genetics and nCounter Core Facility, Heidelberg University, Heidelberg, Germany
| | - Emre Balta
- Institute of Immunology, Section Molecular Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jutta Schröder-Braunstein
- Institute of Immunology, Section Molecular Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Heidelberg University Hospital, Heidelberg, Germany
- *Correspondence: Yvonne Samstag,
| |
Collapse
|
127
|
A bead-based method for high-throughput mapping of the sequence- and force-dependence of T cell activation. Nat Methods 2022; 19:1295-1305. [PMID: 36064771 DOI: 10.1038/s41592-022-01592-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
Adaptive immunity relies on T lymphocytes that use αβ T cell receptors (TCRs) to discriminate among peptides presented by major histocompatibility complex molecules (pMHCs). Identifying pMHCs capable of inducing robust T cell responses will not only enable a deeper understanding of the mechanisms governing immune responses but could also have broad applications in diagnosis and treatment. T cell recognition of sparse antigenic pMHCs in vivo relies on biomechanical forces. However, in vitro screening methods test potential pMHCs without force and often at high (nonphysiological) pMHC densities and thus fail to predict potent agonists in vivo. Here, we present a technology termed BATTLES (biomechanically assisted T cell triggering for large-scale exogenous-pMHC screening) that uses biomechanical force to initiate T cell triggering for peptides and cells in parallel. BATTLES displays candidate pMHCs on spectrally encoded beads composed of a thermo-responsive polymer capable of applying shear loads to T cells, facilitating exploration of the force- and sequence-dependent landscape of T cell responses. BATTLES can be used to explore basic T cell mechanobiology and T cell-based immunotherapies.
Collapse
|
128
|
Cho JH, Tsao WC, Naghizadeh A, Liu D. Standardized protocol for the evaluation of chimeric antigen receptor (CAR)-modified cell immunological synapse quality using the glass-supported planar lipid bilayer. Methods Cell Biol 2022; 173:155-171. [PMID: 36653082 PMCID: PMC10768727 DOI: 10.1016/bs.mcb.2022.07.009] [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] [Indexed: 02/04/2023]
Abstract
Chimeric antigen receptor (CAR)-modified cell therapy is an effective therapy that harnesses the power of the human immune system by re-engineering immune cells that specifically kill tumor cells with tumor antigen specificity. Key to the effective elimination of tumor cells is the establishment of the immunological synapse (IS) between CAR-modified immune cells and their susceptible tumors. For functional activity, CAR-modified cells must form an effective IS to kill tumor cells specifically. The formation of the CAR-specific IS requires the coordination of many cellular processes including reorganization of the cytoskeletal structure, polarization of lytic granules, accumulation of tumor antigen, and phosphorylation of key signaling molecules within the IS. Visualization and assessment of the CAR IS quality can reveal much about the molecular mechanisms that underlie the efficacy of various CAR-modified immune cells. This chapter provides a standardized method of assessing the IS quality by quantifying the tumor antigen (defining the CAR IS formation), cytoskeleton (key component of CAR IS structure), and various molecules of interest involved in the IS formation (key molecular mechanism signatures of CAR IS function) using immunofluorescence on the glass-supported planar lipid bilayer, with a focus on tumor antigen only in this study. We provide specific insights and helpful tips for reagent and sample preparation, assay design, and machine learning (ML)-based data analysis. The protocol described in this chapter will provide a valuable tool to visualize and assess the IS quality of various CAR-modified immune cells.
Collapse
Affiliation(s)
- Jong Hyun Cho
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, United States; Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Wei-Chung Tsao
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, United States; Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Alireza Naghizadeh
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, United States; Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, United States; Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States.
| |
Collapse
|
129
|
Wu Y, Biswas D, Swanton C. Impact of cancer evolution on immune surveillance and checkpoint inhibitor response. Semin Cancer Biol 2022; 84:89-102. [PMID: 33631295 PMCID: PMC9253787 DOI: 10.1016/j.semcancer.2021.02.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/21/2022]
Abstract
Intratumour heterogeneity (ITH) is pervasive across all cancers studied and may provide the evolving tumour multiple routes to escape immune surveillance. Immune checkpoint inhibitors (CPIs) are rapidly becoming standard of care for many cancers. Here, we discuss recent work investigating the influence of ITH on patient response to immune checkpoint inhibitor (CPI) therapy. At its simplest, ITH may confound the diagnostic accuracy of predictive biomarkers used to stratify patients for CPI therapy. Furthermore, ITH is fuelled by mechanisms of genetic instability that can both engage immune surveillance and drive immune evasion. A greater appreciation of the interplay between ITH and the immune system may hold the key to increasing the proportion of patients experiencing durable responses from CPI therapy.
Collapse
Affiliation(s)
- Yin Wu
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, WC1E 6DD, UK; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Dhruva Biswas
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, WC1E 6DD, UK; Bill Lyons Informatics Centre, University College London Cancer Institute, Paul O'Gorman Building, London, WC1E 6DD, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, WC1E 6DD, UK.
| |
Collapse
|
130
|
Oh J, Xia X, Wong WKR, Wong SHD, Yuan W, Wang H, Lai CHN, Tian Y, Ho YP, Zhang H, Zhang Y, Li G, Lin Y, Bian L. The Effect of the Nanoparticle Shape on T Cell Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107373. [PMID: 35297179 DOI: 10.1002/smll.202107373] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/08/2022] [Indexed: 06/14/2023]
Abstract
The mechanism of extracellular ligand nano-geometry in ex vivo T cell activation for immunotherapy remains elusive. Herein, the authors demonstrate large aspect ratio (AR) of gold nanorods (AuNRs) conjugated on cell culture substrate enhancing both murine and human T cell activation through the nanoscale anisotropic presentation of stimulatory ligands (anti-CD3(αCD3) and anti-CD28(αCD28) antibodies). AuNRs with large AR bearing αCD3 and αCD28 antibodies significantly promote T cell expansion and key cytokine secretion including interleukin-2 (IL-2), interferon-gamma (IFN-γ), and tumor necrosis factor-alpha (TNF-α). High membrane tension observed in large AR AuNRs regulates actin filament and focal adhesion assembly and develops maturation-related morphological features in T cells such as membrane ruffle formation, cell spreading, and large T cell receptor (TCR) cluster formation. Anisotropic stimulatory ligand presentation promotes differentiation of naïve CD8+ T cells toward the effector phenotype inducing CD137 expression upon co-culture with human cervical carcinoma. The findings suggest the importance of manipulating extracellular ligand nano-geometry in optimizing T cell behaviors to enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Jiwon Oh
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Xingyu Xia
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, 999077, China
| | - Wai Ki Ricky Wong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Weihao Yuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
- Department of Orthopedic and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, Hong Kong, 999077, China
| | - Haixing Wang
- Department of Orthopedic and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, Hong Kong, 999077, China
| | - Chun Him Nathanael Lai
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Ye Tian
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, 999077, China
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Honglu Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yuan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Gang Li
- Department of Orthopedic and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, Hong Kong, 999077, China
| | - Yuan Lin
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, 999077, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New territories, Hong Kong, 999077, China
- HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, Guang Dong, 518000, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
131
|
Boni N, Shapiro L, Honig B, Wu Y, Rubinstein R. On the formation of ordered protein assemblies in cell-cell interfaces. Proc Natl Acad Sci U S A 2022; 119:e2206175119. [PMID: 35969779 PMCID: PMC9407605 DOI: 10.1073/pnas.2206175119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/20/2022] [Indexed: 11/18/2022] Open
Abstract
Crystal structures of many cell-cell adhesion receptors reveal the formation of linear "molecular zippers" comprising an ordered one-dimensional array of proteins that form both intercellular (trans) and intracellular (cis) interactions. The clustered protocadherins (cPcdhs) provide an exemplar of this phenomenon and use it as a basis of barcoding of vertebrate neurons. Here, we report both Metropolis and kinetic Monte Carlo simulations of cPcdh zipper formation using simplified models of cPcdhs that nevertheless capture essential features of their three-dimensional structure. The simulations reveal that the formation of long zippers is an implicit feature of cPcdh structure and is driven by their cis and trans interactions that have been quantitatively characterized in previous work. Moreover, in agreement with cryo-electron tomography studies, the zippers are found to organize into two-dimensional arrays even in the absence of attractive interactions between individual zippers. Our results suggest that the formation of ordered two-dimensional arrays of linear zippers of adhesion proteins is a common feature of cell-cell interfaces. From the perspective of simulations, they demonstrate the importance of a realistic depiction of adhesion protein structure and interactions if important biological phenomena are to be properly captured.
Collapse
Affiliation(s)
- Nadir Boni
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Lawrence Shapiro
- Zuckerman Mind, Brain and Behavior Institute, Columbia University, New York, NY 10027
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032
| | - Barry Honig
- Zuckerman Mind, Brain and Behavior Institute, Columbia University, New York, NY 10027
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032
- Department of Systems Biology, Columbia University, New York, NY 10032
- Department of Medicine, Division of Nephrology, Columbia University, New York, NY 10032
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Rotem Rubinstein
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
132
|
Cai E, Beppler C, Eichorst J, Marchuk K, Eastman SW, Krummel MF. T cells use distinct topographical and membrane receptor scanning strategies that individually coalesce during receptor recognition. Proc Natl Acad Sci U S A 2022; 119:e2203247119. [PMID: 35914144 PMCID: PMC9372542 DOI: 10.1073/pnas.2203247119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/22/2022] [Indexed: 02/03/2023] Open
Abstract
During immune surveillance, CD8 T cells scan the surface of antigen-presenting cells using dynamic microvillar palpation and movements as well as by having their receptors preconcentrated into patches. Here, we use real-time lattice light-sheet microscopy to demonstrate the independence of microvillar and membrane receptor patch scanning. While T cell receptor (TCR) patches can distribute to microvilli, they do so stochastically and not preferentially as for other receptors such as CD62L. The distinctness of TCR patch movement from microvillar movement extends to many other receptors that form patches that also scan independent of the TCR. An exception to this is the CD8 coreceptor which largely comigrates in patches that overlap with or are closely adjacent to those containing TCRs. Microvilli that assemble into a synapse contain various arrays of the engaged patches, notably of TCRs and the inhibitory receptor PD-1, creating a pastiche of occupancies that vary from microvillar contact to contact. In summary, this work demonstrates that localization of receptor patches within the membrane and on microvillar projections is random prior to antigen detection and that such random variation may play into the generation of many individually composed receptor patch compositions at a single synapse.
Collapse
Affiliation(s)
- En Cai
- Department of Pathology, University of California, San Francisco, CA 94143-0511
| | - Casey Beppler
- Department of Pathology, University of California, San Francisco, CA 94143-0511
| | - John Eichorst
- Department of Pathology, University of California, San Francisco, CA 94143-0511
- Biological Imaging Development CoLab, University of California, San Francisco, CA 94143-0511
| | - Kyle Marchuk
- Department of Pathology, University of California, San Francisco, CA 94143-0511
- Biological Imaging Development CoLab, University of California, San Francisco, CA 94143-0511
- ImmunoX Initiative, University of California, San Francisco, CA 94143-0511
| | - Scott W. Eastman
- Lilly Research Laboratories, Eli Lilly and Company, New York, NY 10016
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, CA 94143-0511
- ImmunoX Initiative, University of California, San Francisco, CA 94143-0511
| |
Collapse
|
133
|
Molon B, Liboni C, Viola A. CD28 and chemokine receptors: Signalling amplifiers at the immunological synapse. Front Immunol 2022; 13:938004. [PMID: 35983040 PMCID: PMC9379342 DOI: 10.3389/fimmu.2022.938004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/08/2022] [Indexed: 01/14/2023] Open
Abstract
T cells are master regulators of the immune response tuning, among others, B cells, macrophages and NK cells. To exert their functions requiring high sensibility and specificity, T cells need to integrate different stimuli from the surrounding microenvironment. A finely tuned signalling compartmentalization orchestrated in dynamic platforms is an essential requirement for the proper and efficient response of these cells to distinct triggers. During years, several studies have depicted the pivotal role of the cytoskeleton and lipid microdomains in controlling signalling compartmentalization during T cell activation and functions. Here, we discuss mechanisms responsible for signalling amplification and compartmentalization in T cell activation, focusing on the role of CD28, chemokine receptors and the actin cytoskeleton. We also take into account the detrimental effect of mutations carried by distinct signalling proteins giving rise to syndromes characterized by defects in T cell functionality.
Collapse
Affiliation(s)
- Barbara Molon
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- *Correspondence: Barbara Molon,
| | - Cristina Liboni
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Antonella Viola
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
134
|
Liang W, Li K, Zhang Q, Li K, Ai K, Zhang J, Jiao X, Li J, Wei X, Yang J. Interleukin-2 inducible T cell kinase (ITK) may participate in the anti-bacterial immune response of Nile tilapia via regulating T-cell activation. FISH & SHELLFISH IMMUNOLOGY 2022; 127:419-426. [PMID: 35779809 DOI: 10.1016/j.fsi.2022.06.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/28/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023]
Abstract
Interleukin-2 inducible T cell kinase (ITK) plays a predominant role in the T-cell receptor (TCR) signaling cascade to ensure valid T-cell activation and function. Nevertheless, whether it regulates T-cell response of early vertebrates remains unknown. Herein, we investigated the involvement of ITK in the lymphocyte-mediated adaptive immune response, and its regulation to T-cell activation in the Nile tilapia Oreochromis niloticus. Both sequence and structure of O. niloticus ITK (OnITK) were remarkably conserved with its homologues from other vertebrates, implying its potential conserved function. OnITK mRNA was extensively expressed in lymphoid-related tissues, and with the relative highest level in peripheral blood. Once Nile tilapia was infected by Edwardsiella piscicida, OnITK in splenic lymphocytes was significantly up-regulated on 7-day post infection at both transcription and translation levels, suggesting that OnITK might involve in the primary adaptive immune response of teleost. Furthermore, upon splenic lymphocytes were stimulated by T-cell specific mitogen PHA, OnITK mRNA and protein levels were dramatically elevated. More importantly, treatment of splenic lymphocytes with specific inhibitor significantly crippled OnITK expression, which in turn impaired the inducible expression of T-cell activation markers IFN-γ, IL-2 and CD122, indicating the critical roles of ITK in regulating T-cell activation of Nile tilapia. Taken together, our results suggest that ITK takes part in the lymphocyte-mediated adaptive immunity of tilapia, and is indispensable for T-cell activation of teleost. Our findings thus provide novel evidences for understanding the mechanism regulating T-cell immunity of early vertebrates, as well as the evolution of adaptive immune system.
Collapse
Affiliation(s)
- Wei Liang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kunming Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kete Ai
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xinying Jiao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiaqi Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| |
Collapse
|
135
|
Kobayashi E, Jin A, Hamana H, Shitaoka K, Tajiri K, Kusano S, Yokoyama S, Ozawa T, Obata T, Muraguchi A, Kishi H. Rapid cloning of antigen-specific T-cell receptors by leveraging the cis activation of T cells. Nat Biomed Eng 2022; 6:806-818. [PMID: 35393565 DOI: 10.1038/s41551-022-00874-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 02/24/2022] [Indexed: 02/08/2023]
Abstract
It is commonly understood that T cells are activated via trans interactions between antigen-specific T-cell receptors (TCRs) and antigenic peptides presented on major histocompatibility complex (MHC) molecules on antigen-presenting cells. By analysing a large number of T cells at the single-cell level on a microwell array, we show that T-cell activation can occur via cis interactions (where TCRs on the T cell interact with the antigenic peptides presented on MHC class-I molecules on the same cell), and that such cis activation can be used to detect antigen-specific T cells and clone their TCR within 4 d. We used the detection-and-cloning system to clone a tumour-antigen-specific TCR from peripheral blood mononuclear cells of healthy donors. TCR cloning by leveraging the cis activation of T cells may facilitate the development of TCR-engineered T cells for cancer therapy.
Collapse
Affiliation(s)
- Eiji Kobayashi
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Aishun Jin
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
- Department of Immunology, Chongqing Medical University, Chongqing, China
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Hiroshi Hamana
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Kiyomi Shitaoka
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
- Department of Immunology, Hiroshima University, Hiroshima, Japan
| | - Kazuto Tajiri
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
- The Third Department of Internal Medicine, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Seisuke Kusano
- RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Japan
| | - Shigeyuki Yokoyama
- RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Japan
| | - Tatsuhiko Ozawa
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Tsutomu Obata
- Toyama Industrial Technology Research and Development Center, Takaoka, Japan
| | - Atsushi Muraguchi
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Hiroyuki Kishi
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan.
| |
Collapse
|
136
|
Dobosz P, Stempor PA, Ramírez Moreno M, Bulgakova NA. Transcriptional and post-transcriptional regulation of checkpoint genes on the tumour side of the immunological synapse. Heredity (Edinb) 2022; 129:64-74. [PMID: 35459932 PMCID: PMC9273643 DOI: 10.1038/s41437-022-00533-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is a disease of the genome, therefore, its development has a clear Mendelian component, demonstrated by well-studied genes such as BRCA1 and BRCA2 in breast cancer risk. However, it is known that a single genetic variant is not enough for cancer to develop leading to the theory of multistage carcinogenesis. In many cases, it is a sequence of events, acquired somatic mutations, or simply polygenic components with strong epigenetic effects, such as in the case of brain tumours. The expression of many genes is the product of the complex interplay between several factors, including the organism's genotype (in most cases Mendelian-inherited), genetic instability, epigenetic factors (non-Mendelian-inherited) as well as the immune response of the host, to name just a few. In recent years the importance of the immune system has been elevated, especially in the light of the immune checkpoint genes discovery and the subsequent development of their inhibitors. As the expression of these genes normally suppresses self-immunoreactivity, their expression by tumour cells prevents the elimination of the tumour by the immune system. These discoveries led to the rapid growth of the field of immuno-oncology that offers new possibilities of long-lasting and effective treatment options. Here we discuss the recent advances in the understanding of the key mechanisms controlling the expression of immune checkpoint genes in tumour cells.
Collapse
Affiliation(s)
- Paula Dobosz
- Central Clinical Hospital of the Ministry of Interior Affairs and Administration in Warsaw, Warsaw, Poland
| | | | - Miguel Ramírez Moreno
- School of Biosciences and Bateson Centre, The University of Sheffield, Sheffield, UK
| | - Natalia A Bulgakova
- School of Biosciences and Bateson Centre, The University of Sheffield, Sheffield, UK.
| |
Collapse
|
137
|
Joshi H, Morley SC. Efficient T Cell Migration and Activation Require L-Plastin. Front Immunol 2022; 13:916137. [PMID: 35844504 PMCID: PMC9277003 DOI: 10.3389/fimmu.2022.916137] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/02/2022] [Indexed: 11/28/2022] Open
Abstract
Rapid re-organization of the actin cytoskeleton supports T-cell trafficking towards immune sites and interaction with antigen presenting cells (APCs). F-actin rearrangement enables T-cell trafficking by stabilizing adhesion to vascular endothelial cells and promoting transendothelial migration. T-cell/APC immune synapse (IS) maturation also relies upon f-actin-anchored LFA-1:ICAM-1 ligation. Therefore, efficient T-cell responses require tight regulation of f-actin dynamics. In this review, we summarize how the actin-bundling protein L-plastin (LPL) regulates T-cell activation and migration. LPL enhances f-actin polymerization and also directly binds to the β2 chain of the integrin LFA-1 to support intercellular adhesion and IS formation in human and murine T cells. LPL- deficient T cells migrate slowly in response to chemo-attractants such as CXCL12, CCL19, and poorly polarize towards ICAM-1. Loss of LPL impairs thymic egress and intranodal motility. LPL is also required for T-cell IS maturation with APCs, and therefore for efficient cytokine production and proliferation. LPL-/- mice are less susceptible to T-cell mediated pathologies, such as allograft rejection and experimental autoimmune encephalomyelitis (EAE). LPL activity is regulated by its N-terminal “headpiece”, which contains serine and threonine phosphorylation and calcium- and calmodulin-binding sites. LPL phosphorylation is required for lamellipodia formation during adhesion and migration, and also for LFA-1 clustering during IS formation. However, the precise molecular interactions by which LPL supports T-cell functional responses remain unclear. Future studies elucidating LPL-mediated regulation of T-cell migration and/or activation may illuminate pathways for therapeutic targeting in T-cell-mediated diseases.
Collapse
Affiliation(s)
- Hemant Joshi
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO, United States
| | - Sharon Celeste Morley
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO, United States
- *Correspondence: Sharon Celeste Morley,
| |
Collapse
|
138
|
Waldman MM, Rahkola JT, Sigler AL, Chung JW, Willett BAS, Kedl RM, Friedman RS, Jacobelli J. Ena/VASP Protein-Mediated Actin Polymerization Contributes to Naïve CD8 + T Cell Activation and Expansion by Promoting T Cell-APC Interactions In Vivo. Front Immunol 2022; 13:856977. [PMID: 35757762 PMCID: PMC9222560 DOI: 10.3389/fimmu.2022.856977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Naïve T cell activation in secondary lymphoid organs such as lymph nodes (LNs) occurs upon recognition of cognate antigen presented by antigen presenting cells (APCs). T cell activation requires cytoskeleton rearrangement and sustained interactions with APCs. Enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) proteins are a family of cytoskeletal effector proteins responsible for actin polymerization and are frequently found at the leading edge of motile cells. Ena/VASP proteins have been implicated in motility and adhesion in various cell types, but their role in primary T cell interstitial motility and activation has not been explored. Our goal was to determine the contribution of Ena/VASP proteins to T cell–APC interactions, T cell activation, and T cell expansion in vivo. Our results showed that naïve T cells from Ena/VASP-deficient mice have a significant reduction in antigen-specific T cell accumulation following Listeria monocytogenes infection. The kinetics of T cell expansion impairment were further confirmed in Ena/VASP-deficient T cells stimulated via dendritic cell immunization. To investigate the cause of this T cell expansion defect, we analyzed T cell–APC interactions in vivo by two-photon microscopy and observed fewer Ena/VASP-deficient naïve T cells interacting with APCs in LNs during priming. We also determined that Ena/VASP-deficient T cells formed conjugates with significantly less actin polymerization at the T cell–APC synapse, and that these conjugates were less stable than their WT counterparts. Finally, we found that Ena/VASP-deficient T cells have less LFA-1 polarized to the T cell–APC synapse. Thus, we conclude that Ena/VASP proteins contribute to T cell actin remodeling during T cell–APC interactions, which promotes the initiation of stable T cell conjugates during APC scanning. Therefore, Ena/VASP proteins are required for efficient activation and expansion of T cells in vivo.
Collapse
Affiliation(s)
- Monique M Waldman
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeremy T Rahkola
- Rocky Mountain Regional Veterans Affairs (VA) Medical Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ashton L Sigler
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeffrey W Chung
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Benjamin A S Willett
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rachel S Friedman
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jordan Jacobelli
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
| |
Collapse
|
139
|
An C, Wang X, Song F, Hu J, Li L. Insights into intercellular receptor-ligand binding kinetics in cell communication. Front Bioeng Biotechnol 2022; 10:953353. [PMID: 35837553 PMCID: PMC9273785 DOI: 10.3389/fbioe.2022.953353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/09/2022] [Indexed: 01/14/2023] Open
Abstract
Cell-cell communication is crucial for cells to sense, respond and adapt to environmental cues and stimuli. The intercellular communication process, which involves multiple length scales, is mediated by the specific binding of membrane-anchored receptors and ligands. Gaining insight into two-dimensional receptor-ligand binding kinetics is of great significance for understanding numerous physiological and pathological processes, and stimulating new strategies in drug design and discovery. To this end, extensive studies have been performed to illuminate the underlying mechanisms that control intercellular receptor-ligand binding kinetics via experiment, theoretical analysis and numerical simulation. It has been well established that the cellular microenvironment where the receptor-ligand interaction occurs plays a vital role. In this review, we focus on the advances regarding the regulatory effects of three factors including 1) protein-membrane interaction, 2) biomechanical force, and 3) bioelectric microenvironment to summarize the relevant experimental observations, underlying mechanisms, as well as their biomedical significances and applications. Meanwhile, we introduce modeling methods together with experiment technologies developed for dealing with issues at different scales. We also outline future directions to advance the field and highlight that building up systematic understandings for the coupling effects of these regulatory factors can greatly help pharmaceutical development.
Collapse
Affiliation(s)
- Chenyi An
- School of Biology and Engineering, Guizhou Medical University, Guiyang, China
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohuan Wang
- Department of Rehabilitation Medicine, Peking University Third Hospital, Beijing, China
| | - Fan Song
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jinglei Hu
- Kuang Yaming Honors School and Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
140
|
Santos F, Valderas-Gutiérrez J, Pérez Del Río E, Castellote-Borrell M, Rodriguez XR, Veciana J, Ratera I, Guasch J. Enhanced human T cell expansion with inverse opal hydrogels. Biomater Sci 2022; 10:3730-3738. [PMID: 35660816 DOI: 10.1039/d2bm00486k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Advanced personalized immunotherapies still have to overcome several biomedical and technical limitations before they become a routine cancer treatment in spite of recent achievements. In adoptive cell therapy (ACT), the capacity to obtain adequate numbers of therapeutic T cells in the patients following ex vivo treatment should be improved. Moreover, the time and costs to produce these T cells should be reduced. In this work, inverse opal (IOPAL) 3D hydrogels consisting of poly(ethylene) glycol (PEG) covalently combined with heparin were engineered to resemble the environment of lymph nodes, where T cells get activated and proliferate. The introduction of an IOPAL strategy allowed a precise control on the porosity of the hydrogels, providing an increase in the proliferation of primary human CD4+ T cells, when compared with state-of-the-art expansion systems. Additionally, the IOPAL hydrogels also showed a superior expansion compared to hydrogels with the same composition, but without the predetermined pore structure. In summary, we have shown the beneficial effect of having an IOPAL architecture in our 3D hydrogels to help achieving large numbers of cells, while maintaining the desired selected phenotypes required for ACT.
Collapse
Affiliation(s)
- Fabião Santos
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, 08193, Spain. .,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Campus UAB, Bellaterra, 08193, Spain
| | | | - Eduardo Pérez Del Río
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, 08193, Spain. .,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Campus UAB, Bellaterra, 08193, Spain
| | - Miquel Castellote-Borrell
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, 08193, Spain. .,Dynamic Biomimetics for Cancer Immunotherapy, Max Planck Partner Group, ICMAB-CSIC, Campus UAB, Bellaterra, 08193, Spain
| | - Xavier Rodriguez Rodriguez
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, 08193, Spain. .,Dynamic Biomimetics for Cancer Immunotherapy, Max Planck Partner Group, ICMAB-CSIC, Campus UAB, Bellaterra, 08193, Spain
| | - Jaume Veciana
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, 08193, Spain. .,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Campus UAB, Bellaterra, 08193, Spain
| | - Imma Ratera
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, 08193, Spain. .,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Campus UAB, Bellaterra, 08193, Spain
| | - Judith Guasch
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, 08193, Spain. .,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Campus UAB, Bellaterra, 08193, Spain.,Dynamic Biomimetics for Cancer Immunotherapy, Max Planck Partner Group, ICMAB-CSIC, Campus UAB, Bellaterra, 08193, Spain
| |
Collapse
|
141
|
Beyond GWAS-Could Genetic Differentiation within the Allograft Rejection Pathway Shape Natural Immunity to COVID-19? Int J Mol Sci 2022; 23:ijms23116272. [PMID: 35682950 PMCID: PMC9181155 DOI: 10.3390/ijms23116272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 01/25/2023] Open
Abstract
COVID-19 infections pose a serious global health concern so it is crucial to identify the biomarkers for the susceptibility to and resistance against this disease that could help in a rapid risk assessment and reliable decisions being made on patients' treatment and their potential hospitalisation. Several studies investigated the factors associated with severe COVID-19 outcomes that can be either environmental, population based, or genetic. It was demonstrated that the genetics of the host plays an important role in the various immune responses and, therefore, there are different clinical presentations of COVID-19 infection. In this study, we aimed to use variant descriptive statistics from GWAS (Genome-Wide Association Study) and variant genomic annotations to identify metabolic pathways that are associated with a severe COVID-19 infection as well as pathways related to resistance to COVID-19. For this purpose, we applied a custom-designed mixed linear model implemented into custom-written software. Our analysis of more than 12.5 million SNPs did not indicate any pathway that was significant for a severe COVID-19 infection. However, the Allograft rejection pathway (hsa05330) was significant (p = 0.01087) for resistance to the infection. The majority of the 27 SNP marking genes constituting the Allograft rejection pathway were located on chromosome 6 (19 SNPs) and the remainder were mapped to chromosomes 2, 3, 10, 12, 20, and X. This pathway comprises several immune system components crucial for the self versus non-self recognition, but also the components of antiviral immunity. Our study demonstrated that not only single variants are important for resistance to COVID-19, but also the cumulative impact of several SNPs within the same pathway matters.
Collapse
|
142
|
LFA1 Activation: Insights from a Single-Molecule Approach. Cells 2022; 11:cells11111751. [PMID: 35681446 PMCID: PMC9179313 DOI: 10.3390/cells11111751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
Integrin LFA1 is a cell adhesion receptor expressed exclusively in leukocytes, and plays crucial roles in lymphocyte trafficking, antigen recognition, and effector functions. Since the discovery that the adhesiveness of LFA1 can be dynamically changed upon stimulation, one challenge has been understanding how integrins are regulated by inside-out signaling coupled with macromolecular conformational changes, as well as ligand bindings that transduce signals from the extracellular domain to the cytoplasm in outside-in signaling. The small GTPase Rap1 and integrin adaptor proteins talin1 and kindlin-3 have been recognized as critical molecules for integrin activation. However, their cooperative regulation of integrin adhesiveness in lymphocytes requires further research. Recent advances in single-molecule imaging techniques have revealed dynamic molecular processes in real-time and provided insight into integrin activation in cellular environments. This review summarizes integrin regulation and discusses new findings regarding the bidirectionality of LFA1 activation and signaling processes in lymphocytes.
Collapse
|
143
|
McKenzie B, Khazen R, Valitutti S. Greek Fire, Poison Arrows, and Scorpion Bombs: How Tumor Cells Defend Against the Siege Weapons of Cytotoxic T Lymphocytes. Front Immunol 2022; 13:894306. [PMID: 35592329 PMCID: PMC9110820 DOI: 10.3389/fimmu.2022.894306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 01/05/2023] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) are the main cellular effectors of the adaptive immune response against cancer cells, which in turn have evolved sophisticated cellular defense mechanisms to withstand CTL attack. Herein we provide a critical review of the pertinent literature on early and late attack/defense events taking place at the CTL/target cell lytic synapse. We examine the earliest steps of CTL-mediated cytotoxicity (“the poison arrows”) elicited within seconds of CTL/target cell encounter, which face commensurately rapid synaptic repair mechanisms on the tumor cell side, providing the first formidable barrier to CTL attack. We examine how breach of this first defensive barrier unleashes the inextinguishable “Greek fire” in the form of granzymes whose broad cytotoxic potential is linked to activation of cell death executioners, injury of vital organelles, and destruction of intracellular homeostasis. Herein tumor cells deploy slower but no less sophisticated defensive mechanisms in the form of enhanced autophagy, increased reparative capacity, and dysregulation of cell death pathways. We discuss how the newly discovered supra-molecular attack particles (SMAPs, the “scorpion bombs”), seek to overcome the robust defensive mechanisms that confer tumor cell resistance. Finally, we discuss the implications of the aforementioned attack/defense mechanisms on the induction of regulated cell death (RCD), and how different contemporary RCD modalities (including apoptosis, pyroptosis, and ferroptosis) may have profound implications for immunotherapy. Thus, we propose that understanding and targeting multiple steps of the attack/defense process will be instrumental to enhance the efficacy of CTL anti-tumor activity and meet the outstanding challenges in clinical immunotherapy.
Collapse
Affiliation(s)
- Brienne McKenzie
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Roxana Khazen
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Salvatore Valitutti
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France.,Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Toulouse, France
| |
Collapse
|
144
|
Shin HG, Yang HR, Yoon A, Lee S. Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy. Int J Mol Sci 2022; 23:5686. [PMID: 35628495 PMCID: PMC9146966 DOI: 10.3390/ijms23105686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second leading cause of death worldwide after cardiovascular diseases. Harnessing the power of immune cells is a promising strategy to improve the antitumor effect of cancer immunotherapy. Recent progress in recombinant DNA technology and antibody engineering has ushered in a new era of bispecific antibody (bsAb)-based immune-cell engagers (ICEs), including T- and natural-killer-cell engagers. Since the first approval of blinatumomab by the United States Food and Drug Administration (US FDA), various bsAb-based ICEs have been developed for the effective treatment of patients with cancer. Simultaneously, several potential therapeutic targets of bsAb-based ICEs have been identified in various cancers. Therefore, this review focused on not only highlighting the action mechanism, design and structure, and status of bsAb-based ICEs in clinical development and their approval by the US FDA for human malignancy treatment, but also on summarizing the currently known and emerging therapeutic targets in cancer. This review provides insights into practical considerations for developing next-generation ICEs.
Collapse
Affiliation(s)
- Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Aerin Yoon
- R&D Division, GC Biopharma, Yongin 16924, Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Korea
| |
Collapse
|
145
|
Jensen LG, Hoh TY, Williamson DJ, Griffié J, Sage D, Rubin-Delanchy P, Owen DM. Correction of multiple-blinking artifacts in photoactivated localization microscopy. Nat Methods 2022; 19:594-602. [PMID: 35545712 DOI: 10.1038/s41592-022-01463-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 03/18/2022] [Indexed: 11/09/2022]
Abstract
Photoactivated localization microscopy (PALM) produces an array of localization coordinates by means of photoactivatable fluorescent proteins. However, observations are subject to fluorophore multiple blinking and each protein is included in the dataset an unknown number of times at different positions, due to localization error. This causes artificial clustering to be observed in the data. We present a 'model-based correction' (MBC) workflow using calibration-free estimation of blinking dynamics and model-based clustering to produce a corrected set of localization coordinates representing the true underlying fluorophore locations with enhanced localization precision, outperforming the state of the art. The corrected data can be reliably tested for spatial randomness or analyzed by other clustering approaches, and descriptors such as the absolute number of fluorophores per cluster are now quantifiable, which we validate with simulated data and experimental data with known ground truth. Using MBC, we confirm that the adapter protein, the linker for activation of T cells, is clustered at the T cell immunological synapse.
Collapse
Affiliation(s)
- Louis G Jensen
- Department of Mathematics, Aarhus University, Aarhus, Denmark.
| | - Tjun Yee Hoh
- Institute for Statistical Science, School of Mathematics, University of Bristol, Bristol, UK
| | - David J Williamson
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Juliette Griffié
- Laboratory of Experimental Biophysics, Institute of Physics, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Daniel Sage
- Biomedical Imaging Group, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Patrick Rubin-Delanchy
- Institute for Statistical Science, School of Mathematics, University of Bristol, Bristol, UK.
| | - Dylan M Owen
- Institute of Immunology and Immunotherapy, School of Mathematics and Centre of Membrane Proteins and Receptors, University of Birmingham, Birmingham, UK.
| |
Collapse
|
146
|
Mahadik R, Kiptoo P, Tolbert T, Siahaan TJ. Immune Modulation by Antigenic Peptides and Antigenic Peptide Conjugates for Treatment of Multiple Sclerosis. MEDICAL RESEARCH ARCHIVES 2022; 10:10.18103/mra.v10i5.2804. [PMID: 36381196 PMCID: PMC9648198 DOI: 10.18103/mra.v10i5.2804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The immune system defends our body by fighting infection from pathogens utilizing both the innate and adaptive immune responses. The innate immune response is generated rapidly as the first line of defense. It is followed by the adaptive immune response that selectively targets infected cells. The adaptive immune response is generated more slowly, but selectively, by targeting a wide range of foreign particles (i.e., viruses or bacteria) or molecules that enter the body, known as antigens. Autoimmune diseases are the results of immune system glitches, where the body's adaptive system recognizes self-antigens as foreign. Thus, the host immune system attacks the self-tissues or organs with a high level of inflammation and causes debilitation in patients. Many current treatments for autoimmune diseases (i.e., multiple sclerosis (MS), rheumatoid arthritis (RA)) have been effective but lead to adverse side effects due to general immune system suppression, which makes patients vulnerable to opportunistic infections. To counter these negative effects, many different avenues of antigen specific treatments are being developed to selectively target the autoreactive immune cells for a specific self-antigen or set of self-antigens while not compromising the general immune system. These approaches include soluble antigenic peptides, bifunctional peptide inhibitors (BPI) including IDAC and Fc-BPI, polymer conjugates, and peptide-drug conjugates. Here, various antigen-specific methods of potential treatments, their efficacy, and limitations will be discussed along with the potential mechanisms of action.
Collapse
Affiliation(s)
- Rucha Mahadik
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, 2093 Constant Avenue, Lawrence, KS 66047
| | | | - Tom Tolbert
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, 2093 Constant Avenue, Lawrence, KS 66047
| | - Teruna J Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, 2093 Constant Avenue, Lawrence, KS 66047
| |
Collapse
|
147
|
Sun J, Zhong X, Fu X, Miller H, Lee P, Yu B, Liu C. The Actin Regulators Involved in the Function and Related Diseases of Lymphocytes. Front Immunol 2022; 13:799309. [PMID: 35371070 PMCID: PMC8965893 DOI: 10.3389/fimmu.2022.799309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/01/2022] [Indexed: 11/21/2022] Open
Abstract
Actin is an important cytoskeletal protein involved in signal transduction, cell structure and motility. Actin regulators include actin-monomer-binding proteins, Wiskott-Aldrich syndrome (WAS) family of proteins, nucleation proteins, actin filament polymerases and severing proteins. This group of proteins regulate the dynamic changes in actin assembly/disassembly, thus playing an important role in cell motility, intracellular transport, cell division and other basic cellular activities. Lymphocytes are important components of the human immune system, consisting of T-lymphocytes (T cells), B-lymphocytes (B cells) and natural killer cells (NK cells). Lymphocytes are indispensable for both innate and adaptive immunity and cannot function normally without various actin regulators. In this review, we first briefly introduce the structure and fundamental functions of a variety of well-known and newly discovered actin regulators, then we highlight the role of actin regulators in T cell, B cell and NK cell, and finally provide a landscape of various diseases associated with them. This review provides new directions in exploring actin regulators and promotes more precise and effective treatments for related diseases.
Collapse
Affiliation(s)
- Jianxuan Sun
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyu Zhong
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Fu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Bing Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
148
|
Dai Z, Mu W, Zhao Y, Cheng J, Lin H, Ouyang K, Jia X, Liu J, Wei Q, Wang M, Liu C, Tan T, Zhou J. T cells expressing CD5/CD7 bispecific chimeric antigen receptors with fully human heavy-chain-only domains mitigate tumor antigen escape. Signal Transduct Target Ther 2022; 7:85. [PMID: 35332132 PMCID: PMC8948246 DOI: 10.1038/s41392-022-00898-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/11/2022] [Accepted: 01/19/2022] [Indexed: 12/23/2022] Open
Abstract
Bispecific chimeric antigen receptor T-cell (CAR-T) therapies have shown promising results in clinical trials for advanced B-cell malignancies. However, it is challenging to broaden the success of bispecific CAR-T therapies to treat refractory/relapse (r/r) T-cell leukemia/lymphoma because targeting multiple T-cell-expressing antigens leads to exacerbated CAR-T cell fratricide and potential safety concerns. Fully human heavy chain variable (FHVH) antibodies that specifically target CD5 or CD7 were screened and constructed to CD5/CD7 bispecific CARs. A truncated Epidermal growth factor receptor were integrated into CAR constructs to address safety concerns. To tackle the fratricidal issue of CAR-T cells targeting T-cell-pan marker(s), CRISPR/Cas9-based CD5 and CD7 genes knockout were performed before lentiviral transduction of bispecific CARs. Functional comparison between different bispecific CAR structures: tandem CARs and dual CAR were performed in vitro and in vivo to determine the optimal construct suitable for addressing T-cell malignancy antigen escape in clinical setting. Knockout of CD5 and CD7 prevents fratricide of CD5/CD7 bispecific CAR-T cells, and FHVH-derived CD5/CD7 bispecific CAR-T cells demonstrate potent antitumor activity in vitro and in vivo. The fratricide-resistant FHVH-derived CD5/CD7 bispecific CAR-T cells have potent antitumor activity against T-cell malignancies, and tandem CARs are more effective than dual CAR in preventing tumor escape in heterogeneous leukemic cells. The meaningful clinical efficacy and safety of tandem CD5/CD7 CAR-T cells deserve to be explored urgently.
Collapse
Affiliation(s)
- Zhenyu Dai
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ya Zhao
- Nanjing IASO Biotherapeutics, Nanjing, Jiangsu, 210000, China
| | - Jiali Cheng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Haolong Lin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Kedong Ouyang
- Nanjing IASO Biotherapeutics, Nanjing, Jiangsu, 210000, China
| | - Xiangyin Jia
- Nanjing IASO Biotherapeutics, Nanjing, Jiangsu, 210000, China
| | - Jianwei Liu
- Nanjing IASO Biotherapeutics, Nanjing, Jiangsu, 210000, China
| | - Qiaoe Wei
- Nanjing IASO Biotherapeutics, Nanjing, Jiangsu, 210000, China
| | - Meng Wang
- Nanjing IASO Biotherapeutics, Nanjing, Jiangsu, 210000, China
| | - Chaohong Liu
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Taochao Tan
- Nanjing IASO Biotherapeutics, Nanjing, Jiangsu, 210000, China.
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
149
|
Abstract
Immune signalling pathways convert pathogenic stimuli into cytosolic events that lead to the resolution of infection. Upon ligand engagement, immune receptors together with their downstream adaptors and effectors undergo substantial conformational changes and spatial reorganization. During this process, nanometre-to-micrometre-sized signalling clusters have been commonly observed that are believed to be hotspots for signal transduction. Because of their large size and heterogeneous composition, it remains a challenge to fully understand the mechanisms by which these signalling clusters form and their functional consequences. Recently, phase separation has emerged as a new biophysical principle for organizing biomolecules into large clusters with fluidic properties. Although the field is still in its infancy, studies of phase separation in immunology are expected to provide new perspectives for understanding immune responses. Here, we present an up-to-date view of how liquid-liquid phase separation drives the formation of signalling condensates and regulates immune signalling pathways, including those downstream of T cell receptor, B cell receptor and the innate immune receptors cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and retinoic acid-inducible gene I protein (RIG-I). We conclude with a summary of the current challenges the field is facing and outstanding questions for future studies.
Collapse
Affiliation(s)
- Qian Xiao
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - Ceara K McAtee
- Yale Combined Program in the Biological and Biomedical Sciences, New Haven, CT, USA
| | - Xiaolei Su
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, Yale University, New Haven, CT, USA.
| |
Collapse
|
150
|
Arias CF, Acosta FJ, Fernandez-Arias C. Killing the competition: a theoretical framework for liver-stage malaria. Open Biol 2022; 12:210341. [PMID: 35350863 PMCID: PMC8965401 DOI: 10.1098/rsob.210341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The first stage of malaria infections takes place inside the host's hepatocytes. Remarkably, Plasmodium parasites do not infect hepatocytes immediately after reaching the liver. Instead, they migrate through several hepatocytes before infecting their definitive host cells, thus increasing their chances of immune destruction. Considering that malaria can proceed normally without cell traversal, this is indeed a puzzling behaviour. In fact, the role of hepatocyte traversal remains unknown to date, implying that the current understanding of malaria is incomplete. In this work, we hypothesize that the parasites traverse hepatocytes to actively trigger an immune response in the host. This behaviour would be part of a strategy of superinfection exclusion aimed to reduce intraspecific competition during the blood stage of the infection. Based on this hypothesis, we formulate a comprehensive theory of liver-stage malaria that integrates all the available knowledge about the infection. The interest of this new paradigm is not merely theoretical. It highlights major issues in the current empirical approach to the study of Plasmodium and suggests new strategies to fight malaria.
Collapse
Affiliation(s)
- Clemente F. Arias
- Centro de Investigaciones Biológicas (CSIC), Madrid, Spain,Grupo Interdisciplinar de Sistemas Complejos de Madrid, Spain
| | | | - Cristina Fernandez-Arias
- Departamento de Inmunología, Universidad Complutense de Madrid, Spain,Instituto de Medicina Molecular, Universidade de Lisboa, Portugal
| |
Collapse
|