151
|
Naghizadeh A, Tsao WC, Hyun Cho J, Xu H, Mohamed M, Li D, Xiong W, Metaxas D, Ramos CA, Liu D. In vitro machine learning-based CAR T immunological synapse quality measurements correlate with patient clinical outcomes. PLoS Comput Biol 2022; 18:e1009883. [PMID: 35303007 PMCID: PMC8955962 DOI: 10.1371/journal.pcbi.1009883] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 01/28/2022] [Indexed: 12/04/2022] Open
Abstract
The human immune system consists of a highly intelligent network of billions of independent, self-organized cells that interact with each other. Machine learning (ML) is an artificial intelligence (AI) tool that automatically processes huge amounts of image data. Immunotherapies have revolutionized the treatment of blood cancer. Specifically, one such therapy involves engineering immune cells to express chimeric antigen receptors (CAR), which combine tumor antigen specificity with immune cell activation in a single receptor. To improve their efficacy and expand their applicability to solid tumors, scientists optimize different CARs with different modifications. However, predicting and ranking the efficacy of different "off-the-shelf" immune products (e.g., CAR or Bispecific T-cell Engager [BiTE]) and selection of clinical responders are challenging in clinical practice. Meanwhile, identifying the optimal CAR construct for a researcher to further develop a potential clinical application is limited by the current, time-consuming, costly, and labor-intensive conventional tools used to evaluate efficacy. Particularly, more than 30 years of immunological synapse (IS) research data demonstrate that T cell efficacy is not only controlled by the specificity and avidity of the tumor antigen and T cell interaction, but also it depends on a collective process, involving multiple adhesion and regulatory molecules, as well as tumor microenvironment, spatially and temporally organized at the IS formed by cytotoxic T lymphocytes (CTL) and natural killer (NK) cells. The optimal function of cytotoxic lymphocytes (including CTL and NK) depends on IS quality. Recognizing the inadequacy of conventional tools and the importance of IS in immune cell functions, we investigate a new strategy for assessing CAR-T efficacy by quantifying CAR IS quality using the glass-support planar lipid bilayer system combined with ML-based data analysis. Previous studies in our group show that CAR-T IS quality correlates with antitumor activities in vitro and in vivo. However, current manually quantified IS quality data analysis is time-consuming and labor-intensive with low accuracy, reproducibility, and repeatability. In this study, we develop a novel ML-based method to quantify thousands of CAR cell IS images with enhanced accuracy and speed. Specifically, we used artificial neural networks (ANN) to incorporate object detection into segmentation. The proposed ANN model extracts the most useful information to differentiate different IS datasets. The network output is flexible and produces bounding boxes, instance segmentation, contour outlines (borders), intensities of the borders, and segmentations without borders. Based on requirements, one or a combination of this information is used in statistical analysis. The ML-based automated algorithm quantified CAR-T IS data correlates with the clinical responder and non-responder treated with Kappa-CAR-T cells directly from patients. The results suggest that CAR cell IS quality can be used as a potential composite biomarker and correlates with antitumor activities in patients, which is sufficiently discriminative to further test the CAR IS quality as a clinical biomarker to predict response to CAR immunotherapy in cancer. For translational research, the method developed here can also provide guidelines for designing and optimizing numerous CAR constructs for potential clinical development. Trial Registration: ClinicalTrials.gov NCT00881920.
Collapse
Affiliation(s)
- Alireza Naghizadeh
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, New Jersey, United States of America
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, United States of America
| | - Wei-chung Tsao
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, New Jersey, United States of America
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, United States of America
| | - Jong Hyun Cho
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, New Jersey, United States of America
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, United States of America
| | - Hongye Xu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, New Jersey, United States of America
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, United States of America
| | - Mohab Mohamed
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, New Jersey, United States of America
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, United States of America
| | - Dali Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Wei Xiong
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Dimitri Metaxas
- Department of Computer Science, Rutgers University, Piscataway Township, New Jersey, United States of America
| | - Carlos A. Ramos
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, New Jersey, United States of America
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, United States of America
| |
Collapse
|
152
|
Dam T, Chouliara M, Junghans V, Jönsson P. Supported Lipid Bilayers and the Study of Two-Dimensional Binding Kinetics. Front Mol Biosci 2022; 9:833123. [PMID: 35252352 PMCID: PMC8896763 DOI: 10.3389/fmolb.2022.833123] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Binding between protein molecules on contacting cells is essential in initiating and regulating several key biological processes. In contrast to interactions between molecules in solution, these events are restricted to the two-dimensional (2D) plane of the meeting cell surfaces. However, converting between the more commonly available binding kinetics measured in solution and the so-called 2D binding kinetics has proven a complicated task since for the latter several factors other than the protein-protein interaction per se have an impact. A few important examples of these are: protein density, membrane fluctuations, force on the bond and the use of auxiliary binding molecules. The development of model membranes, and in particular supported lipid bilayers (SLBs), has made it possible to simplify the studied contact to analyze these effects and to measure 2D binding kinetics of individual protein-protein interactions. We will in this review give an overview of, and discuss, how different SLB systems have been used for this and compare different methods to measure binding kinetics in cell-SLB contacts. Typically, the SLB is functionalized with fluorescently labelled ligands whose interaction with the corresponding receptor on a binding cell can be detected. This interaction can either be studied 1) by an accumulation of ligands in the cell-SLB contact, whose magnitude depends on the density of the proteins and binding affinity of the interaction, or 2) by tracking single ligands in the SLB, which upon interaction with a receptor result in a change of motion of the diffusing ligand. The advantages and disadvantages of other methods measuring 2D binding kinetics will also be discussed and compared to the fluorescence-based methods. Although binding kinetic measurements in cell-SLB contacts have provided novel information on how ligands interact with receptors in vivo the number of these measurements is still limited. This is influenced by the complexity of the system as well as the required experimental time. Moreover, the outcome can vary significantly between studies, highlighting the necessity for continued development of methods to study 2D binding kinetics with higher precision and ease.
Collapse
Affiliation(s)
- Tommy Dam
- Department of Chemistry, Lund University, Lund, Sweden
| | | | - Victoria Junghans
- Nuffield Department of Medicine, CAMS Oxford Institute, University of Oxford, Oxford, United Kingdom
| | - Peter Jönsson
- Department of Chemistry, Lund University, Lund, Sweden
- *Correspondence: Peter Jönsson,
| |
Collapse
|
153
|
Greco B, Malacarne V, De Girardi F, Scotti GM, Manfredi F, Angelino E, Sirini C, Camisa B, Falcone L, Moresco MA, Paolella K, Di Bono M, Norata R, Sanvito F, Arcangeli S, Doglioni C, Ciceri F, Bonini C, Graziani A, Bondanza A, Casucci M. Disrupting N-glycan expression on tumor cells boosts chimeric antigen receptor T cell efficacy against solid malignancies. Sci Transl Med 2022; 14:eabg3072. [PMID: 35044789 DOI: 10.1126/scitranslmed.abg3072] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Immunotherapy with chimeric antigen receptor (CAR)-engineered T cells showed exceptional successes in patients with refractory B cell malignancies. However, first-in-human studies in solid tumors revealed unique hurdles contributing to poor demonstration of efficacy. Understanding the determinants of tumor recognition by CAR T cells should translate into the design of strategies that can overcome resistance. Here, we show that multiple carcinomas express extracellular N-glycans, whose abundance negatively correlates with CAR T cell killing. By knocking out mannoside acetyl-glucosaminyltransferase 5 (MGAT5) in pancreatic adenocarcinoma (PAC), we showed that N-glycans protect tumors from CAR T cell killing by interfering with proper immunological synapse formation and reducing transcriptional activation, cytokine production, and cytotoxicity. To overcome this barrier, we exploited the high metabolic demand of tumors to safely inhibit N-glycans synthesis with the glucose/mannose analog 2-deoxy-d-glucose (2DG). Treatment with 2DG disrupts the N-glycan cover on tumor cells and results in enhanced CAR T cell activity in different xenograft mouse models of PAC. Moreover, 2DG treatment interferes with the PD-1-PD-L1 axis and results in a reduced exhaustion profile of tumor-infiltrating CAR T cells in vivo. The combined 2DG and CAR T cell therapy was successful against multiple carcinomas besides PAC, including those arising from the lung, ovary, and bladder, and with different clinically relevant CAR specificities, such as CD44v6 and CEA. Overall, our results indicate that tumor N-glycosylation regulates the quality and magnitude of CAR T cell responses, paving the way for the rational design of improved therapies against solid malignancies.
Collapse
Affiliation(s)
- Beatrice Greco
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Valeria Malacarne
- Lipid Signaling in Cancer and Metabolism Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10124 Torino, Italy
| | - Federica De Girardi
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulia Maria Scotti
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Elia Angelino
- Lipid Signaling in Cancer and Metabolism Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10124 Torino, Italy
| | - Camilla Sirini
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Barbara Camisa
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Laura Falcone
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marta Angiola Moresco
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Katia Paolella
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mattia Di Bono
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Rossana Norata
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesca Sanvito
- Pathology Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Silvia Arcangeli
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudio Doglioni
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Pathology Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Hematology and Hematopoietic Stem Cell Transplantation Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Chiara Bonini
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Graziani
- Lipid Signaling in Cancer and Metabolism Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10124 Torino, Italy
| | - Attilio Bondanza
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Monica Casucci
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
154
|
Proteomic analysis in primary T cells reveals IL-7 alters T cell receptor thresholding via CYTIP/cytohesin/LFA-1 localisation and activation. Biochem J 2022; 479:225-243. [PMID: 35015072 PMCID: PMC8883493 DOI: 10.1042/bcj20210313] [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: 05/11/2021] [Revised: 12/15/2021] [Accepted: 01/11/2022] [Indexed: 11/17/2022]
Abstract
The ability of the cellular immune system to discriminate self from foreign antigens depends on the appropriate calibration of the T cell receptor (TCR) signalling threshold. The lymphocyte homeostatic cytokine interleukin 7 (IL-7) is known to affect TCR thresholding, but the molecular mechanism is not fully elucidated. A better understanding of this process is highly relevant in the context of autoimmune disease therapy and cancer immunotherapy. We sought to characterise the early signalling events attributable to IL-7 priming; in particular, the altered phosphorylation of signal transduction proteins and their molecular localisation to the TCR. By integrating high-resolution proximity- phospho-proteomic and imaging approaches using primary T cells, rather than engineered cell lines or an in vitro expanded T cell population, we uncovered transduction events previously not linked to IL-7. We show that IL-7 leads to dephosphorylation of cytohesin interacting protein (CYTIP) at a hitherto undescribed phosphorylation site (pThr280) and alters the co-localisation of cytohesin-1 with the TCR and LFA-1 integrin. These results show that IL-7, acting via CYTIP and cytohesin-1, may impact TCR activation thresholds by enhancing the co-clustering of TCR and LFA-1 integrin.
Collapse
|
155
|
Al-Aghbar MA, Jainarayanan AK, Dustin ML, Roffler SR. The interplay between membrane topology and mechanical forces in regulating T cell receptor activity. Commun Biol 2022; 5:40. [PMID: 35017678 PMCID: PMC8752658 DOI: 10.1038/s42003-021-02995-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
T cells are critically important for host defense against infections. T cell activation is specific because signal initiation requires T cell receptor (TCR) recognition of foreign antigen peptides presented by major histocompatibility complexes (pMHC) on antigen presenting cells (APCs). Recent advances reveal that the TCR acts as a mechanoreceptor, but it remains unclear how pMHC/TCR engagement generates mechanical forces that are converted to intracellular signals. Here we propose a TCR Bending Mechanosignal (TBM) model, in which local bending of the T cell membrane on the nanometer scale allows sustained contact of relatively small pMHC/TCR complexes interspersed among large surface receptors and adhesion molecules on the opposing surfaces of T cells and APCs. Localized T cell membrane bending is suggested to increase accessibility of TCR signaling domains to phosphorylation, facilitate selective recognition of agonists that form catch bonds, and reduce noise signals associated with slip bonds.
Collapse
Affiliation(s)
- Mohammad Ameen Al-Aghbar
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Ashwin K Jainarayanan
- Interdisciplinary Bioscience Doctoral Training Program and Exeter College, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
156
|
T cell activation and effector function in the human Jurkat T cell model. Methods Cell Biol 2022. [PMID: 37516527 DOI: 10.1016/bs.mcb.2022.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In order to understand T cell function, it is necessary to completely decipher the molecular dynamics underlying T cell activation and effector function. In vitro easy-to-handle cellular models are valuable tools to study intracellular molecular mechanisms in live cells. The CD4 T cell line Jurkat (JK) has been widely employed to investigate intracellular signaling leading to T cell activation in response to T cell receptor (TCR) triggering. Here, we describe diverse, complementary protocols to evaluate the TCR- and costimulation-mediated T cell activation, as well as the immunological synapse assembly and cytokine production occurring as a consequence of successful early activation events. This in vitro model is extremely useful to address molecular mechanisms operating during T cell activation and effector function acting in diverse pathophysiological scenarios.
Collapse
|
157
|
Wang H, Shang J, He Z, Zheng M, Jia H, Zhang Y, Yang W, Gao X, Gao F. Dual peptide nanoparticles platform for enhanced antigen-specific immune tolerance for treatment of experimental autoimmune encephalomyelitis. Biomater Sci 2022; 10:3878-3891. [DOI: 10.1039/d2bm00444e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Current therapeutic strategies for autoimmune diseases including multiple sclerosis (MS) are directed toward nonspecific immunosuppression which has severe side effects. The induction of antigen-specific tolerance becomes an ideal therapy for...
Collapse
|
158
|
Kawashima M, Higuchi H, Kotani A. Significance of trogocytosis and exosome-mediated transport in establishing and maintaining the tumor microenvironment in lymphoid malignancies. J Clin Exp Hematop 2021; 61:192-201. [PMID: 34193756 PMCID: PMC8808107 DOI: 10.3960/jslrt.21005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/05/2021] [Accepted: 04/04/2021] [Indexed: 11/25/2022] Open
Abstract
It is widely accepted that the tumor microenvironment plays an important role in the progression of lymphoid malignancies. Interaction between the tumor and its surrounding immune cells is considered a potential therapeutic target. For example, anti-programmed cell death 1 (PD-1) antibody stimulates the surrounding exhausted immune cells to release PD-1/PD-L1, thereby leading to the regression of PD-L1-positive tumors. Recently, biological phenomena, such as trogocytosis and exosome-mediated transport were demonstrated to be involved in establishing and maintaining the tumor microenvironment. We found that trogocytosis-mediated PD-L1/L2 transfer from tumor cells to monocytes/macrophages is involved in immune dysfunction in classic Hodgkin lymphoma. Exosomes derived from Epstein-Barr virus (EBV)-associated lymphoma cells induce lymphoma tumorigenesis by transferring the EBV-coding microRNAs from the infected cells to macrophages. In this review, we summarized these biological phenomena based on our findings.
Collapse
|
159
|
Li W, Li F, Zhang X, Lin HK, Xu C. Insights into the post-translational modification and its emerging role in shaping the tumor microenvironment. Signal Transduct Target Ther 2021; 6:422. [PMID: 34924561 PMCID: PMC8685280 DOI: 10.1038/s41392-021-00825-8] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/11/2022] Open
Abstract
More and more in-depth studies have revealed that the occurrence and development of tumors depend on gene mutation and tumor heterogeneity. The most important manifestation of tumor heterogeneity is the dynamic change of tumor microenvironment (TME) heterogeneity. This depends not only on the tumor cells themselves in the microenvironment where the infiltrating immune cells and matrix together forming an antitumor and/or pro-tumor network. TME has resulted in novel therapeutic interventions as a place beyond tumor beds. The malignant cancer cells, tumor infiltrate immune cells, angiogenic vascular cells, lymphatic endothelial cells, cancer-associated fibroblastic cells, and the released factors including intracellular metabolites, hormonal signals and inflammatory mediators all contribute actively to cancer progression. Protein post-translational modification (PTM) is often regarded as a degradative mechanism in protein destruction or turnover to maintain physiological homeostasis. Advances in quantitative transcriptomics, proteomics, and nuclease-based gene editing are now paving the global ways for exploring PTMs. In this review, we focus on recent developments in the PTM area and speculate on their importance as a critical functional readout for the regulation of TME. A wealth of information has been emerging to prove useful in the search for conventional therapies and the development of global therapeutic strategies.
Collapse
Affiliation(s)
- Wen Li
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China
| | - Feifei Li
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Chuan Xu
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China.
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA.
| |
Collapse
|
160
|
Carreras-Sureda A, Abrami L, Ji-Hee K, Wang WA, Henry C, Frieden M, Didier M, van der Goot FG, Demaurex N. S-acylation by ZDHHC20 targets ORAI1 channels to lipid rafts for efficient Ca 2+ signaling by Jurkat T cell receptors at the immune synapse. eLife 2021; 10:72051. [PMID: 34913437 PMCID: PMC8683079 DOI: 10.7554/elife.72051] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/29/2021] [Indexed: 12/29/2022] Open
Abstract
Efficient immune responses require Ca2+ fluxes across ORAI1 channels during engagement of T cell receptors (TCR) at the immune synapse (IS) between T cells and antigen presenting cells. Here, we show that ZDHHC20-mediated S-acylation of the ORAI1 channel at residue Cys143 promotes TCR recruitment and signaling at the IS. Cys143 mutations reduced ORAI1 currents and store-operated Ca2+ entry in HEK-293 cells and nearly abrogated long-lasting Ca2+ elevations, NFATC1 translocation, and IL-2 secretion evoked by TCR engagement in Jurkat T cells. The acylation-deficient channel remained in cholesterol-poor domains upon enforced ZDHHC20 expression and was recruited less efficiently to the IS along with actin and TCR. Our results establish S-acylation as a critical regulator of ORAI1 channel trafficking and function at the IS and reveal that ORAI1 S-acylation enhances TCR recruitment to the synapse.
Collapse
Affiliation(s)
| | - Laurence Abrami
- Faculty of Life Sciences, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Kim Ji-Hee
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Wen-An Wang
- Department of Cell Physiology and Metabolism, Geneva, Switzerland
| | | | - Maud Frieden
- Department of Cell Physiology and Metabolism, Geneva, Switzerland
| | - Monica Didier
- Department of Cell Physiology and Metabolism, Geneva, Switzerland
| | - F Gisou van der Goot
- Faculty of Life Sciences, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, Geneva, Switzerland
| |
Collapse
|
161
|
Shah K, Al-Haidari A, Sun J, Kazi JU. T cell receptor (TCR) signaling in health and disease. Signal Transduct Target Ther 2021; 6:412. [PMID: 34897277 PMCID: PMC8666445 DOI: 10.1038/s41392-021-00823-w] [Citation(s) in RCA: 246] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Interaction of the T cell receptor (TCR) with an MHC-antigenic peptide complex results in changes at the molecular and cellular levels in T cells. The outside environmental cues are translated into various signal transduction pathways within the cell, which mediate the activation of various genes with the help of specific transcription factors. These signaling networks propagate with the help of various effector enzymes, such as kinases, phosphatases, and phospholipases. Integration of these disparate signal transduction pathways is done with the help of adaptor proteins that are non-enzymatic in function and that serve as a scaffold for various protein-protein interactions. This process aids in connecting the proximal to distal signaling pathways, thereby contributing to the full activation of T cells. This review provides a comprehensive snapshot of the various molecules involved in regulating T cell receptor signaling, covering both enzymes and adaptors, and will discuss their role in human disease.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Amr Al-Haidari
- Clinical Genetics and Pathology, Skåne University Hospital, Region Skåne, Lund, Sweden
- Clinical Sciences Department, Surgery Research Unit, Lund University, Malmö, Sweden
| | - Jianmin Sun
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
162
|
Thapa P, Guyer RS, Yang AY, Parks CA, Brusko TM, Brusko M, Connors TJ, Farber DL. Infant T cells are developmentally adapted for robust lung immune responses through enhanced T cell receptor signaling. Sci Immunol 2021; 6:eabj0789. [PMID: 34890254 PMCID: PMC8765725 DOI: 10.1126/sciimmunol.abj0789] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Infants require coordinated immune responses to prevent succumbing to multiple infectious challenges during early life, particularly in the respiratory tract. The mechanisms by which infant T cells are functionally adapted for these responses are not well understood. Here, we demonstrated using an in vivo mouse cotransfer model that infant T cells generated greater numbers of lung-homing effector cells in response to influenza infection compared with adult T cells in the same host, due to augmented T cell receptor (TCR)–mediated signaling. Mouse infant T cells showed increased sensitivity to low antigen doses, originating at the interface between T cells and antigen-bearing accessory cells—through actin-mediated mobilization of signaling molecules to the immune synapse. This enhanced signaling was also observed in human infant versus adult T cells. Our findings provide a mechanism for how infants control pathogen load and dissemination, which is important for designing developmentally targeted strategies for promoting immune responses at this vulnerable life stage.
Collapse
Affiliation(s)
- Puspa Thapa
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York NY 10032
| | - Rebecca S. Guyer
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York NY 10032
| | - Alexander Y. Yang
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York NY 10032
| | - Christopher A. Parks
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY 10032
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032
| | - Todd M. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611
| | - Maigan Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611
| | - Thomas J. Connors
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032
| | - Donna L. Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York NY 10032
- Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032
| |
Collapse
|
163
|
Li L, Gao J, Shao Y, Song F, Hu J. Tuning cell adhesion on supported lipid bilayers via nanoscale geometry. SOFT MATTER 2021; 17:10376-10382. [PMID: 34747961 DOI: 10.1039/d1sm01407b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The cell-supported lipid bilayer (SLB) adhesion system has been widely used as the model system to study the receptor-ligand interactions that occur at the membrane interface. The ligand-functionalized SLBs are deposited either directly on solids or on polymer cushions. An important question that arises is whether the geometry of the SLB affects the binding of cell adhesion receptors to the ligands. By using a mesoscopic mechanical model and Monte Carlo simulations, we have investigated the adhesion of a fluid membrane to a corrugated or egg-carton shaped SLB. We find that the nanoscale geometry of the SLB strongly affects the receptor-ligand binding. This effect results from the fact that the adhering membrane bends according to the SLB geometry in order for the adhesion receptors to bind ligands. The membrane bending couples with spatial distribution of the receptor-ligand complexes and membrane thermal undulations. Our results demonstrate that cell adhesion to SLBs can be controlled by tuning the nanoscale geometry of the SLB, and may have profound implications for future development of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- 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.
| | - Jie Gao
- Kuang Yaming Honors School and Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Yingfeng Shao
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 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.
| |
Collapse
|
164
|
Chen H, Smith M, Herz J, Li T, Hasley R, Le Saout C, Zhu Z, Cheng J, Gronda A, Martina JA, Irusta PM, Karpova T, McGavern DB, Catalfamo M. The role of protease-activated receptor 1 signaling in CD8 T cell effector functions. iScience 2021; 24:103387. [PMID: 34841225 PMCID: PMC8605340 DOI: 10.1016/j.isci.2021.103387] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
CD8 T cells are essential for adaptive immunity against viral infections. Protease activated receptor 1 (PAR1) is expressed by CD8 T cells; however, its role in T cell effector function is not well defined. Here we show that in human CD8 T cells, PAR1 stimulation accelerates calcium mobilization. Furthermore, PAR1 is involved in cytotoxic T cell function by facilitating granule trafficking via actin polymerization and repositioning of the microtubule organizing center (MTOC) toward the immunological synapse. In vivo, PAR1-/- mice have reduced cytokine-producing T cells in response to a lymphocytic choriomeningitis virus (LCMV) infection and fail to efficiently control the virus. Specific deletion of PAR1 in LCMV GP33-specific CD8 T cells results in reduced expansion and diminished effector function. These data demonstrate that PAR1 plays a role in T cell activation and function, and this pathway could represent a new therapeutic strategy to modulate CD8 T cell effector function.
Collapse
Affiliation(s)
- Hui Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mindy Smith
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jasmin Herz
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tong Li
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Rebecca Hasley
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cecile Le Saout
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ziang Zhu
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Jie Cheng
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Andres Gronda
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - José A. Martina
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pablo M. Irusta
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - Tatiana Karpova
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| |
Collapse
|
165
|
Chouliara M, Junghans V, Dam T, Santos AM, Davis SJ, Jönsson P. Single-cell measurements of two-dimensional binding affinity across cell contacts. Biophys J 2021; 120:5032-5040. [PMID: 34653390 PMCID: PMC8633712 DOI: 10.1016/j.bpj.2021.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/21/2021] [Accepted: 10/08/2021] [Indexed: 12/21/2022] Open
Abstract
The two-dimensional (2D) affinity between protein molecules across contacting cells is a key parameter regulating and initiating several cellular processes. However, measuring 2D affinity can be challenging, and experimental data are limited. In addition, the obtained 2D affinities are typically averaged over the cell population. We here present a method to measure 2D affinity on single cells binding to polyhistidine-tagged fluorescent ligands anchored to a supported lipid bilayer (SLB). By decreasing the density of ligands in the SLB using imidazole, a new steady-state accumulation in the contact is obtained, and from this change, both the 2D affinity and the number of receptors on the cell can be determined. The method was validated on an SLB containing rat CD2 binding to the rat CD48 mutant T92A expressed on Jurkat T cells. The addition of imidazole did not influence the average 2D affinity (1/Kd), and the spread in affinities within the cell population was low, Kd = 4.9 ± 0.9 molecules/μm2 (mean ± SD), despite an order of magnitude spread in ligand accumulation because of differences in receptor density. It was also found that cell contact size increased both with ligand density and with the number of receptors per cell but that the contact size stayed approximately constant when lowering the ligand density, above a density of around 10 rat CD2 molecules/μm2, after the contact first had formed, indicative of a heterogeneous process. In summary, this method not only allows for single-cell affinities to be measured, but it can also reduce measurement and analysis time and improve measurement accuracy. Because of the low spread in 2D Kd within the cell population, the analysis can further be restricted to the cells showing the strongest binding, paving the way for using this method to study weak binding events.
Collapse
Affiliation(s)
| | - Victoria Junghans
- Department of Chemistry, Lund University, Lund, Sweden; Radcliffe Department of Medicine and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Tommy Dam
- Department of Chemistry, Lund University, Lund, Sweden
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Simon J Davis
- Radcliffe Department of Medicine and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Peter Jönsson
- Department of Chemistry, Lund University, Lund, Sweden.
| |
Collapse
|
166
|
Velas L, Brameshuber M, Huppa JB, Kurz E, Dustin ML, Zelger P, Jesacher A, Schütz GJ. Three-Dimensional Single Molecule Localization Microscopy Reveals the Topography of the Immunological Synapse at Isotropic Precision below 15 nm. NANO LETTERS 2021; 21:9247-9255. [PMID: 34709845 PMCID: PMC8587899 DOI: 10.1021/acs.nanolett.1c03160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
T-cells engage with antigen-presenting cells in search for antigenic peptides and form transient interfaces termed immunological synapses. Synapse topography affects receptor binding rates and the mutual segregation of proteins due to size exclusion effects. It is hence important to determine the 3D topography of the immunological synapse at high precision. Current methods provide only rather coarse images of the protein distribution within the synapse. Here, we applied supercritical angle fluorescence microscopy combined with defocused imaging, which allows three-dimensional single molecule localization microscopy (3D-SMLM) at an isotropic localization precision below 15 nm. Experiments were performed on hybrid synapses between primary T-cells and functionalized glass-supported lipid bilayers. We used 3D-SMLM to quantify the cleft size within the synapse by mapping the position of the T-cell receptor (TCR) with respect to the supported lipid bilayer, yielding average distances of 18 nm up to 31 nm for activating and nonactivating bilayers, respectively.
Collapse
Affiliation(s)
- Lukas Velas
- Institute
of Applied Physics, TU Wien, 1040 Vienna, Austria
| | | | - Johannes B. Huppa
- Institute
for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology
and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Elke Kurz
- Kennedy
Institute of Rheumatology, University of
Oxford, OX3 7FY Oxford, United Kingdom
| | - Michael L. Dustin
- Kennedy
Institute of Rheumatology, University of
Oxford, OX3 7FY Oxford, United Kingdom
| | - Philipp Zelger
- Division
for Biomedical Physics, Medical University
of Innsbruck, 6020 Innsbruck, Austria
| | - Alexander Jesacher
- Division
for Biomedical Physics, Medical University
of Innsbruck, 6020 Innsbruck, Austria
| | | |
Collapse
|
167
|
Moser T, Hoepner L, Schwenker K, Seiberl M, Feige J, Akgün K, Haschke-Becher E, Ziemssen T, Sellner J. Cladribine Alters Immune Cell Surface Molecules for Adhesion and Costimulation: Further Insights to the Mode of Action in Multiple Sclerosis. Cells 2021; 10:cells10113116. [PMID: 34831335 PMCID: PMC8618022 DOI: 10.3390/cells10113116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/28/2022] Open
Abstract
Cladribine (CLAD) is a deoxyadenosine analogue prodrug which is given in multiple sclerosis (MS) as two short oral treatment courses 12 months apart. Reconstitution of adaptive immune function following selective immune cell depletion is the presumed mode of action. In this exploratory study, we investigated the impact of CLAD tablets on immune cell surface molecules for adhesion (CAMs) and costimulation (CoSs) in people with MS (pwMS). We studied 18 pwMS who started treatment with CLAD and 10 healthy controls (HCs). Peripheral blood mononuclear cells were collected at baseline and every 3 months throughout a 24-month period. We analysed ICAM-1, LFA-1, CD28, HLADR, CD154, CD44, VLA-4 (CD49d/CD29), PSGL-1 and PD-1 with regard to their expression on B and T cells (T helper (Th) and cytotoxic T cells (cT)) and surface density (mean fluorescence intensity, MFI) by flow cytometry. The targeted analysis of CAM and CoS on the surface of immune cells in pwMS revealed a higher percentage of ICAM-1 (B cells, Th, cT), LFA-1 (B cells, cT), HLADR (B cells, cT), CD28 (cT) and CD154 (Th). In pwMS, we found lower frequencies of Th and cT cells expressing PSGL-1 and B cells for the inhibitory signal PD-1, whereas the surface expression of LFA-1 on cT and of HLADR on B cells was denser. Twenty-four months after the first CLAD cycle, the frequencies of B cells expressing CD44, CD29 and CD49d were lower compared with the baseline, together with decreased densities of ICAM-1, CD44 and HLADR. The rate of CD154 expressing Th cells dropped at 12 months. For cT, no changes were seen for frequency or density. Immune reconstitution by oral CLAD was associated with modification of the pro-migratory and -inflammatory surface patterns of CAMs and CoSs in immune cell subsets. This observation pertains primarily to B cells, which are key cells underlying MS pathogenesis.
Collapse
Affiliation(s)
- Tobias Moser
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
- Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (L.H.); (K.A.); (T.Z.)
| | - Lena Hoepner
- Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (L.H.); (K.A.); (T.Z.)
| | - Kerstin Schwenker
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
| | - Michael Seiberl
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
| | - Julia Feige
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
| | - Katja Akgün
- Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (L.H.); (K.A.); (T.Z.)
| | | | - Tjalf Ziemssen
- Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (L.H.); (K.A.); (T.Z.)
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, 80333 München, Germany
- Department of Neurology, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
- Correspondence: ; Tel.: +43-2572-9004-12850; Fax: +43-2572-9004-49281
| |
Collapse
|
168
|
Singh M, Thakur M, Mishra M, Yadav M, Vibhuti R, Menon AM, Nagda G, Dwivedi VP, Dakal TC, Yadav V. Gene regulation of intracellular adhesion molecule-1 (ICAM-1): A molecule with multiple functions. Immunol Lett 2021; 240:123-136. [PMID: 34715236 DOI: 10.1016/j.imlet.2021.10.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 01/04/2023]
Abstract
Intracellular adhesion molecule 1 (ICAM-1) is one of the most extensively studied inducible cell adhesion molecules which is responsible for several immune functions like T cell activation, extravasation, inflammation, etc. The molecule is constitutively expressed over the cell surface and is regulated up / down in response to inflammatory mediators like cellular stress, proinflammatory cytokines, viral infection. These stimuli modulate the expression of ICAM-1 primarily through regulating the ICAM-1 gene transcription. On account of the presence of various binding sites for NF-κB, AP-1, SP-1, and many other transcription factors, the architecture of the ICAM-1 promoter become complex. Transcription factors in union with other transcription factors, coactivators, and suppressors promote their assembly in a stereospecific manner on ICAM-1 promoter which mediates ICAM-1 regulation in response to different stimuli. Along with transcriptional regulation, epigenetic modifications also play a pivotal role in controlling ICAM-1 expression on different cell types. In this review, we summarize the regulation of ICAM-1 expression both at the transcriptional as well as post-transcriptional level with an emphasis on transcription factors and signaling pathways involved.
Collapse
Affiliation(s)
- Mona Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067 India
| | - Mony Thakur
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| | - Manish Mishra
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research- Institute of Microbial Technology, Chandigarh-160036 India
| | - Manisha Yadav
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research- Institute of Microbial Technology, Chandigarh-160036 India
| | - Rajkamal Vibhuti
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| | - Athira M Menon
- Genome and computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001 India
| | - Girima Nagda
- Department of Zoology, Mohanlal Sukhadia University, Udaipur, Rajasthan-313001 India
| | - Ved Prakash Dwivedi
- International Centre for Genetic Engineering and Biotechnology, ICGEB Campus, Aruna Asaf Ali Marg, New Delhi-110067 India
| | - Tikam Chand Dakal
- Genome and computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001 India
| | - Vinod Yadav
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| |
Collapse
|
169
|
Siokis A, Robert PA, Demetriou P, Kvalvaag A, Valvo S, Mayya V, Dustin ML, Meyer-Hermann M. Characterization of mechanisms positioning costimulatory complexes in immune synapses. iScience 2021; 24:103100. [PMID: 34622155 PMCID: PMC8479700 DOI: 10.1016/j.isci.2021.103100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/12/2021] [Accepted: 09/07/2021] [Indexed: 11/30/2022] Open
Abstract
Small immunoglobulin superfamily (sIGSF) adhesion complexes form a corolla of microdomains around an integrin ring and secretory core during immunological synapse (IS) formation. The corolla recruits and retains major costimulatory/checkpoint complexes, such as CD28, making forces that govern corolla formation of particular interest. Here, we investigated the mechanisms underlying molecular reorganization of CD2, an adhesion and costimulatory molecule of the sIGSF family during IS formation. Computer simulations showed passive distal exclusion of CD2 complexes under weak interactions with the ramified F-actin transport network. Attractive forces between CD2 and CD28 complexes relocate CD28 from the IS center to the corolla. Size-based sorting interactions with large glycocalyx components, such as CD45, or short-range CD2 self-attraction successfully explain the corolla 'petals.' This establishes a general simulation framework for complex pattern formation observed in cell-bilayer and cell-cell interfaces, and the suggestion of new therapeutic targets, where boosting or impairing characteristic pattern formation can be pivotal.
Collapse
Affiliation(s)
- Anastasios Siokis
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
| | - Philippe A. Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
| | - Philippos Demetriou
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Audun Kvalvaag
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Salvatore Valvo
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Viveka Mayya
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Michael L. Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig 38106, Germany
| |
Collapse
|
170
|
Feher K, Graus MS, Coelho S, Farrell MV, Goyette J, Gaus K. K-Neighbourhood Analysis: A Method for Understanding SMLM Images as Compositions of Local Neighbourhoods. FRONTIERS IN BIOINFORMATICS 2021; 1:724127. [PMID: 36303786 PMCID: PMC9581049 DOI: 10.3389/fbinf.2021.724127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 10/04/2021] [Indexed: 11/30/2022] Open
Abstract
Single molecule localisation microscopy (SMLM) is a powerful tool that has revealed the spatial arrangement of cell surface signalling proteins, producing data of enormous complexity. The complexity is partly driven by the convolution of technical and biological signal components, and partly by the challenge of pooling information across many distinct cells. To address these two particular challenges, we have devised a novel algorithm called K-neighbourhood analysis (KNA), which emphasises the fact that each image can also be viewed as a composition of local neighbourhoods. KNA is based on a novel transformation, spatial neighbourhood principal component analysis (SNPCA), which is defined by the PCA of the normalised K-nearest neighbour vectors of a spatially random point pattern. Here, we use KNA to define a novel visualisation of individual images, to compare within and between groups of images and to investigate the preferential patterns of phosphorylation. This methodology is also highly flexible and can be used to augment existing clustering methods by providing clustering diagnostics as well as revealing substructure within microclusters. In summary, we have presented a highly flexible analysis tool that presents new conceptual possibilities in the analysis of SMLM images.
Collapse
Affiliation(s)
- Kristen Feher
- School of Medical Sciences, EMBL Australia Node in Single Molecule Science, University of New South Wales, Kensington, NSW, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Matthew S. Graus
- School of Medical Sciences, EMBL Australia Node in Single Molecule Science, University of New South Wales, Kensington, NSW, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Simao Coelho
- School of Medical Sciences, EMBL Australia Node in Single Molecule Science, University of New South Wales, Kensington, NSW, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Megan V. Farrell
- School of Medical Sciences, EMBL Australia Node in Single Molecule Science, University of New South Wales, Kensington, NSW, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Jesse Goyette
- School of Medical Sciences, EMBL Australia Node in Single Molecule Science, University of New South Wales, Kensington, NSW, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Katharina Gaus
- School of Medical Sciences, EMBL Australia Node in Single Molecule Science, University of New South Wales, Kensington, NSW, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
171
|
Bohmwald K, Gálvez NMS, Andrade CA, Mora VP, Muñoz JT, González PA, Riedel CA, Kalergis AM. Modulation of Adaptive Immunity and Viral Infections by Ion Channels. Front Physiol 2021; 12:736681. [PMID: 34690811 PMCID: PMC8531258 DOI: 10.3389/fphys.2021.736681] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022] Open
Abstract
Most cellular functions require of ion homeostasis and ion movement. Among others, ion channels play a crucial role in controlling the homeostasis of anions and cations concentration between the extracellular and intracellular compartments. Calcium (Ca2+) is one of the most relevant ions involved in regulating critical functions of immune cells, allowing the appropriate development of immune cell responses against pathogens and tumor cells. Due to the importance of Ca2+ in inducing the immune response, some viruses have evolved mechanisms to modulate intracellular Ca2+ concentrations and the mobilization of this cation through Ca2+ channels to increase their infectivity and to evade the immune system using different mechanisms. For instance, some viral infections require the influx of Ca2+ through ionic channels as a first step to enter the cell, as well as their replication and budding. Moreover, through the expression of viral proteins on the surface of infected cells, Ca2+ channels function can be altered, enhancing the pathogen evasion of the adaptive immune response. In this article, we review those ion channels and ion transporters that are essential for the function of immune cells. Specifically, cation channels and Ca2+ channels in the context of viral infections and their contribution to the modulation of adaptive immune responses.
Collapse
Affiliation(s)
- Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M. S. Gálvez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina P. Mora
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José T. Muñoz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
172
|
Mastrogiovanni M, Di Bartolo V, Alcover A. Cell Polarity Regulators, Multifunctional Organizers of Lymphocyte Activation and Function. Biomed J 2021; 45:299-309. [PMID: 34626864 PMCID: PMC9250085 DOI: 10.1016/j.bj.2021.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/01/2021] [Accepted: 10/01/2021] [Indexed: 11/27/2022] Open
Abstract
Cell polarity regulators are ubiquitous, evolutionary conserved multifunctional proteins. They contain a variety of protein–protein interaction domains endowing them the capacity to interact with cytoskeleton structures, membrane components and multiple regulatory proteins. In this way, they act in complexes and are pivotal for cell growth and differentiation, tissue formation, stability and turnover, cell migration, wound healing, and others. Hence some of these proteins are tumor suppressors. These cellular processes rely on the establishment of cell polarity characterized by the asymmetric localization of proteins, RNAs, membrane domains, or organelles that together condition cell shape and function. Whether apparently stable, as in epithelia or neurons, or very dynamic, as in immune cells, cell polarity is an active process. It involves cytoskeleton reorganization and targeted intracellular traffic, and results in cellular events such as protein synthesis, secretion and assembly taking place at defined cell poles. Multiple polarity regulators orchestrate these processes. Immune cells are particularly versatile in rapidly polarizing and assuming different shapes, so to swiftly adopt specialized behaviors and functions. Polarity regulators act in various ways in different immune cell types and at their distinct differentiation states. Here we review how cell polarity regulators control different processes and functions along T lymphocyte physiology, including cell migration through different tissues, immunological synapse formation and effector functions.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Lymphocyte Cell Biology Unit, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, Department of Immunology, Institut Pasteur, INSERM-U1224. F-75015 Paris, France; Sorbonne Université, Collège Doctoral, F-75005 Paris. France
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, Department of Immunology, Institut Pasteur, INSERM-U1224. F-75015 Paris, France
| | - Andrés Alcover
- Lymphocyte Cell Biology Unit, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, Department of Immunology, Institut Pasteur, INSERM-U1224. F-75015 Paris, France.
| |
Collapse
|
173
|
Nanoconfinement of microvilli alters gene expression and boosts T cell activation. Proc Natl Acad Sci U S A 2021; 118:2107535118. [PMID: 34599101 DOI: 10.1073/pnas.2107535118] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2021] [Indexed: 12/11/2022] Open
Abstract
T cells sense and respond to their local environment at the nanoscale by forming small actin-rich protrusions, called microvilli, which play critical roles in signaling and antigen recognition, particularly at the interface with the antigen presenting cells. However, the mechanism by which microvilli contribute to cell signaling and activation is largely unknown. Here, we present a tunable engineered system that promotes microvilli formation and T cell signaling via physical stimuli. We discovered that nanoporous surfaces favored microvilli formation and markedly altered gene expression in T cells and promoted their activation. Mechanistically, confinement of microvilli inside of nanopores leads to size-dependent sorting of membrane-anchored proteins, specifically segregating CD45 phosphatases and T cell receptors (TCR) from the tip of the protrusions when microvilli are confined in 200-nm pores but not in 400-nm pores. Consequently, formation of TCR nanoclustered hotspots within 200-nm pores allows sustained and augmented signaling that prompts T cell activation even in the absence of TCR agonists. The synergistic combination of mechanical and biochemical signals on porous surfaces presents a straightforward strategy to investigate the role of microvilli in T cell signaling as well as to boost T cell activation and expansion for application in the growing field of adoptive immunotherapy.
Collapse
|
174
|
Hao J, Zhou H, Nemes K, Yen D, Zhao W, Bramlett C, Wang B, Lu R, Shen K. Membrane-bound SCF and VCAM-1 synergistically regulate the morphology of hematopoietic stem cells. J Cell Biol 2021; 220:212562. [PMID: 34402812 PMCID: PMC8374872 DOI: 10.1083/jcb.202010118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 06/29/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Membrane-bound factors expressed by niche stromal cells constitute a unique class of localized cues and regulate the long-term functions of adult stem cells, yet little is known about the underlying mechanisms. Here, we used a supported lipid bilayer (SLB) to recapitulate the membrane-bound interactions between hematopoietic stem cells (HSCs) and niche stromal cells. HSCs cluster membrane-bound stem cell factor (mSCF) at the HSC-SLB interface. They further form a polarized morphology with aggregated mSCF under a large protrusion through a synergy with VCAM-1 on the bilayer, which drastically enhances HSC adhesion. These features are unique to mSCF and HSCs among the factors and hematopoietic populations we examined. The mSCF-VCAM-1 synergy and the polarized HSC morphology require PI3K signaling and cytoskeletal reorganization. The synergy also enhances nuclear retention of FOXO3a, a crucial factor for HSC maintenance, and minimizes its loss induced by soluble SCF. Our work thus reveals a unique role and signaling mechanism of membrane-bound factors in regulating stem cell morphology and function.
Collapse
Affiliation(s)
- Jia Hao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Kristen Nemes
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Daniel Yen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Winfield Zhao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Charles Bramlett
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA
| | - Bowen Wang
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA
| | - Rong Lu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA.,Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA.,Department of Medicine, University of Southern California, Los Angeles, CA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA.,USC Stem Cell, University of Southern California, Los Angeles, CA
| |
Collapse
|
175
|
Zhou Q, Gu H, Sun S, Zhang Y, Hou Y, Li C, Zhao Y, Ma P, Lv L, Aji S, Sun S, Wang X, Zhan L. Large-Sized Graphene Oxide Nanosheets Increase DC-T-Cell Synaptic Contact and the Efficacy of DC Vaccines against SARS-CoV-2. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102528. [PMID: 34396603 PMCID: PMC8420123 DOI: 10.1002/adma.202102528] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/29/2021] [Indexed: 05/03/2023]
Abstract
Dendritic cell (DC) vaccines are used for cancer and infectious diseases, albeit with limited efficacy. Modulating the formation of DC-T-cell synapses may greatly increase their efficacy. The effects of graphene oxide (GO) nanosheets on DCs and DC-T-cell synapse formation are evaluated. In particular, size-dependent interactions are observed between GO nanosheets and DCs. GOs with diameters of >1 µm (L-GOs) demonstrate strong adherence to the DC surface, inducing cytoskeletal reorganization via the RhoA-ROCK-MLC pathway, while relatively small GOs (≈500 nm) are predominantly internalized by DCs. Furthermore, L-GO treatment enhances DC-T-cell synapse formation via cytoskeleton-dependent membrane positioning of integrin ICAM-1. L-GO acts as a "nanozipper," facilitating the aggregation of DC-T-cell clusters to produce a stable microenvironment for T cell activation. Importantly, L-GO-adjuvanted DCs promote robust cytotoxic T cell immune responses against SARS-CoV-2 spike 1, leading to >99.7% viral RNA clearance in mice infected with a clinically isolated SARS-CoV-2 strain. These findings highlight the potential value of nanomaterials as DC vaccine adjuvants for modulating DC-T-cell synapse formation and provide a basis for the development of effective COVID-19 vaccines.
Collapse
Affiliation(s)
- Qianqian Zhou
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
| | - Hongjing Gu
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAcademy of Military Medical SciencesBeijing100071China
| | - Sujing Sun
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
| | - Yulong Zhang
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
| | - Yangyang Hou
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
| | - Chenyan Li
- BGI collegeZhengzhou UniversityHenan Institute of Medical and Pharmaceutical ScienceZhengzhou UniversityZhengzhou450001P. R. China
| | - Yan Zhao
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
| | - Ping Ma
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
| | - Liping Lv
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
| | - Subi Aji
- Cold Spring Biotech CorporationBeijing110000P. R. China
| | - Shihui Sun
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAcademy of Military Medical SciencesBeijing100071China
| | - Xiaohui Wang
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
| | - Linsheng Zhan
- Institute of Health Service and Transfusion MedicineBeijing100850P. R. China
- BGI collegeZhengzhou UniversityHenan Institute of Medical and Pharmaceutical ScienceZhengzhou UniversityZhengzhou450001P. R. China
| |
Collapse
|
176
|
Wilhelm KB, Morita S, McAffee DB, Kim S, O'Dair MK, Groves JT. Height, but not binding epitope, affects the potency of synthetic TCR agonists. Biophys J 2021; 120:3869-3880. [PMID: 34453921 PMCID: PMC8511163 DOI: 10.1016/j.bpj.2021.08.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/27/2022] Open
Abstract
Under physiological conditions, peptide-major histocompatibility complex (pMHC) molecules can trigger T cell receptors (TCRs) as monovalent ligands that are sparsely distributed on the plasma membrane of an antigen-presenting cell. TCRs can also be triggered by artificial clustering, such as with pMHC tetramers or antibodies; however, these strategies circumvent many of the natural ligand discrimination mechanisms of the T cell and can elicit nonphysiological signaling activity. We have recently introduced a synthetic TCR agonist composed of an anti-TCRβ Fab′ antibody fragment covalently bound to a DNA oligonucleotide, which serves as a membrane anchor. This Fab′-DNA ligand efficiently triggers TCR as a monomer when membrane associated and exhibits a potency and activation profile resembling agonist pMHC. In this report, we explore the geometric requirements for efficient TCR triggering and cellular activation by Fab′-DNA ligands. We find that T cells are insensitive to the ligand binding epitope on the TCR complex but that length of the DNA tether is important. Increasing, the intermembrane distance spanned by Fab′-DNA:TCR complexes decreases TCR triggering efficiency and T cell activation potency, consistent with the kinetic-segregation model of TCR triggering. These results establish design parameters for constructing synthetic TCR agonists that are able to activate polyclonal T cell populations, such as T cells from a human patient, in a similar manner as the native pMHC ligand.
Collapse
Affiliation(s)
- Kiera B Wilhelm
- Department of Chemistry, University of California, Berkeley, California
| | - Shumpei Morita
- Department of Chemistry, University of California, Berkeley, California
| | - Darren B McAffee
- Department of Chemistry, University of California, Berkeley, California
| | - Sungi Kim
- Department of Chemistry, University of California, Berkeley, California
| | - Mark K O'Dair
- Department of Chemistry, University of California, Berkeley, California
| | - Jay T Groves
- Department of Chemistry, University of California, Berkeley, California.
| |
Collapse
|
177
|
Leithner A, Altenburger LM, Hauschild R, Assen FP, Rottner K, Stradal TEB, Diz-Muñoz A, Stein JV, Sixt M. Dendritic cell actin dynamics control contact duration and priming efficiency at the immunological synapse. J Cell Biol 2021; 220:211749. [PMID: 33533935 PMCID: PMC7863705 DOI: 10.1083/jcb.202006081] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 11/25/2020] [Accepted: 01/12/2021] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) are crucial for the priming of naive T cells and the initiation of adaptive immunity. Priming is initiated at a heterologous cell–cell contact, the immunological synapse (IS). While it is established that F-actin dynamics regulates signaling at the T cell side of the contact, little is known about the cytoskeletal contribution on the DC side. Here, we show that the DC actin cytoskeleton is decisive for the formation of a multifocal synaptic structure, which correlates with T cell priming efficiency. DC actin at the IS appears in transient foci that are dynamized by the WAVE regulatory complex (WRC). The absence of the WRC in DCs leads to stabilized contacts with T cells, caused by an increase in ICAM1-integrin–mediated cell–cell adhesion. This results in lower numbers of activated and proliferating T cells, demonstrating an important role for DC actin in the regulation of immune synapse functionality.
Collapse
Affiliation(s)
- Alexander Leithner
- Institute of Science and Technology Austria, Klosterneuburg, Austria.,Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Lukas M Altenburger
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Robert Hauschild
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Frank P Assen
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Klemens Rottner
- Zoological Institute, Technical University Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Units, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Michael Sixt
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| |
Collapse
|
178
|
Lankipalli S, H S MS, Selvam D, Samanta D, Nair D, Ramagopal UA. Cryptic association of B7-2 molecules and its implication for clustering. Protein Sci 2021; 30:1958-1973. [PMID: 34191384 PMCID: PMC8376414 DOI: 10.1002/pro.4151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
T-cell co-stimulation through CD28/CTLA4:B7-1/B7-2 axis is one of the extensively studied pathways that resulted in the discovery of several FDA-approved drugs for autoimmunity and cancer. However, many aspects of the signaling mechanism remain elusive, including oligomeric association and clustering of B7-2 on the cell surface. Here, we describe the structure of the IgV domain of B7-2 and its cryptic association into 1D arrays that appear to represent the pre-signaling state of B7-2 on the cell membrane. Super-resolution microscopy experiments on heterologous cells expressing B7-2 and B7-1 suggest, B7-2 form relatively elongated and larger clusters compared to B7-1. The sequence and structural comparison of other B7 family members, B7-1:CTLA4 and B7-2:CTLA-4 complex structures, support our view that the observed B7-2 1D zipper array is physiologically important. This observed 1D zipper-like array also provides an explanation for its clustering, and upright orientation on the cell surface, and avoidance of spurious signaling.
Collapse
Affiliation(s)
- Swetha Lankipalli
- Biological Sciences DivisionPoornaprajna Institute of Scientific Research (PPISR)BengaluruIndia
- Manipal Academy of Higher EducationManipalKarnatakaIndia
| | | | - Deepak Selvam
- Jawaharlal Nehru Center for Advance Scientific ResearchBengaluruKarnatakaIndia
- National Institute for Research in TuberculosisChennaiIndia
| | - Dibyendu Samanta
- School of Bioscience, Sir J. C. Bose Laboratory ComplexIndian Institute of Technology KharagpurKharagpurIndia
| | - Deepak Nair
- Centre for NeuroscienceIndian Institute of ScienceBangaloreIndia
| | - Udupi A. Ramagopal
- Biological Sciences DivisionPoornaprajna Institute of Scientific Research (PPISR)BengaluruIndia
| |
Collapse
|
179
|
Eidell KP, Lovy A, Sylvain NR, Scangarello FA, Muendlein HI, Ophir MJ, Nguyen K, Seminario MC, Bunnell SC. LFA-1 and kindlin-3 enable the collaborative transport of SLP-76 microclusters by myosin and dynein motors. J Cell Sci 2021; 134:270974. [PMID: 34279667 DOI: 10.1242/jcs.258602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/13/2021] [Indexed: 01/10/2023] Open
Abstract
Integrin engagement within the immune synapse enhances T cell activation, but our understanding of this process is incomplete. In response to T cell receptor (TCR) ligation, SLP-76 (LCP2), ADAP (FYB1) and SKAP55 (SKAP1) are recruited into microclusters and activate integrins via the effectors talin-1 and kindlin-3 (FERMT3). We postulated that integrins influence the centripetal transport and signaling of SLP-76 microclusters via these linkages. We show that contractile myosin filaments surround and are co-transported with SLP-76 microclusters, and that TCR ligand density governs the centripetal movement of both structures. Centripetal transport requires formin activity, actomyosin contraction, microtubule integrity and dynein motor function. Although immobilized VLA-4 (α4β1 integrin) and LFA-1 (αLβ2 integrin) ligands arrest the centripetal movement of SLP-76 microclusters and myosin filaments, VLA-4 acts distally, while LFA-1 acts in the lamellum. Integrin β2, kindlin-3 and zyxin are required for complete centripetal transport, while integrin β1 and talin-1 are not. CD69 upregulation is similarly dependent on integrin β2, kindlin-3 and zyxin, but not talin-1. These findings highlight the integration of cytoskeletal systems within the immune synapse and reveal extracellular ligand-independent roles for LFA-1 and kindlin-3. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Keith P Eidell
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Alenka Lovy
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Nicholas R Sylvain
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Frank A Scangarello
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Hayley I Muendlein
- Graduate Program in Genetics, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Michael J Ophir
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Ken Nguyen
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | | | - Stephen C Bunnell
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
180
|
Johansen KH, Golec DP, Thomsen JH, Schwartzberg PL, Okkenhaug K. PI3K in T Cell Adhesion and Trafficking. Front Immunol 2021; 12:708908. [PMID: 34421914 PMCID: PMC8377255 DOI: 10.3389/fimmu.2021.708908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
PI3K signalling is required for activation, differentiation, and trafficking of T cells. PI3Kδ, the dominant PI3K isoform in T cells, has been extensively characterised using PI3Kδ mutant mouse models and PI3K inhibitors. Furthermore, characterisation of patients with Activated PI3K Delta Syndrome (APDS) and mouse models with hyperactive PI3Kδ have shed light on how increased PI3Kδ activity affects T cell functions. An important function of PI3Kδ is that it acts downstream of TCR stimulation to activate the major T cell integrin, LFA-1, which controls transendothelial migration of T cells as well as their interaction with antigen-presenting cells. PI3Kδ also suppresses the cell surface expression of CD62L and CCR7 which controls the migration of T cells across high endothelial venules in the lymph nodes and S1PR1 which controls lymph node egress. Therefore, PI3Kδ can control both entry and exit of T cells from lymph nodes as well as the recruitment to and retention of T cells within inflamed tissues. This review will focus on the regulation of adhesion receptors by PI3Kδ and how this contributes to T cell trafficking and localisation. These findings are relevant for our understanding of how PI3Kδ inhibitors may affect T cell redistribution and function.
Collapse
Affiliation(s)
- Kristoffer H Johansen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.,Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, United States
| | - Dominic P Golec
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, United States
| | - Julie H Thomsen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
181
|
Rodríguez-Fernández JL, Criado-García O. The Actin Cytoskeleton at the Immunological Synapse of Dendritic Cells. Front Cell Dev Biol 2021; 9:679500. [PMID: 34409027 PMCID: PMC8366227 DOI: 10.3389/fcell.2021.679500] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/05/2021] [Indexed: 01/02/2023] Open
Abstract
Dendritic cells (DCs) are considered the most potent antigen-presenting cells. DCs control the activation of T cells (TCs) in the lymph nodes. This process involves forming a specialized superstructure at the DC-TC contact zone called the immunological synapse (IS). For the sake of clarity, we call IS(DC) and IS(TC) the DC and TC sides of the IS, respectively. The IS(DC) and IS(TC) seem to organize as multicentric signaling hubs consisting of surface proteins, including adhesion and costimulatory molecules, associated with cytoplasmic components, which comprise cytoskeletal proteins and signaling molecules. Most of the studies on the IS have focused on the IS(TC), and the information on the IS(DC) is still sparse. However, the data available suggest that both IS sides are involved in the control of TC activation. The IS(DC) may govern activities of DCs that confer them the ability to activate the TCs. One key component of the IS(DC) is the actin cytoskeleton. Herein, we discuss experimental data that support the concept that actin polarized at the IS(DC) is essential to maintaining IS stability necessary to induce TC activation.
Collapse
Affiliation(s)
- José Luis Rodríguez-Fernández
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | |
Collapse
|
182
|
Gérard A, Cope AP, Kemper C, Alon R, Köchl R. LFA-1 in T cell priming, differentiation, and effector functions. Trends Immunol 2021; 42:706-722. [PMID: 34266767 PMCID: PMC10734378 DOI: 10.1016/j.it.2021.06.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/19/2022]
Abstract
The integrin LFA-1 is crucial for T cell entry into mammalian lymph nodes and tissues, and for promoting interactions with antigen-presenting cells (APCs). However, it is increasingly evident that LFA-1 has additional key roles beyond the mere support of adhesion between T cells, the endothelium, and/or APCs. These include roles in homotypic T cell-T cell (T-T) communication, the induction of intracellular complement activity underlying Th1 effector cell polarization, and the support of long-lasting T cell memory. Here, we briefly summarize current knowledge of LFA-1 biology, discuss novel cytoskeletal regulators of LFA-1 functions, and review new aspects of LFA-1 mechanobiology that are relevant to its function in immunological synapses and in specific pathologies arising from LFA-1 dysregulation.
Collapse
Affiliation(s)
- Audrey Gérard
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew P Cope
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, UK
| | - Claudia Kemper
- National Heart, Lung and Blood Institute (NHLBI), National Institute of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, USA; Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ronen Alon
- The Weizmann Institute of Science, Rehovot, Israel
| | - Robert Köchl
- Peter Gorer Department of Immunobiology, King's College London, London, UK.
| |
Collapse
|
183
|
Liu Y, Bockermann R, Hadi M, Safari I, Carrion B, Kveiborg M, Issazadeh-Navikas S. ADAM12 is a costimulatory molecule that determines Th1 cell fate and mediates tissue inflammation. Cell Mol Immunol 2021; 18:1904-1919. [PMID: 32572163 PMCID: PMC8322154 DOI: 10.1038/s41423-020-0486-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/31/2020] [Indexed: 12/18/2022] Open
Abstract
A disintegrin and metalloproteinase (ADAM)12 was previously found to be expressed in T cells in the inflamed brain. However, the function of ADAM12 in T-cell responses in general and in tissue inflammation has not been examined. Here, we studied the role of ADAM12 in T-cell responses, fate determination on activation, and its functions in T cells to mediate tissue inflammation. We identified ADAM12 as a costimulatory molecule that is expressed on naive T cells and downregulated on stimulation. ADAM12 mimics CD28 costimulatory signaling to activate and induce the proliferation of T-helper 1 (Th1) cells. Monoclonal ADAM12 Fab antibodies trigger T-cell activation by amplifying TCR signaling to stimulate T-bet-mediated IFNγ production. Lack of genomic ADAM12 and its knockdown in T cells diminished T-bet and IFNγ production in Th1 cells, whereas other T cells, including Th17 cells, were unaffected. ADAM12 had similar functions in vivo on myelin antigen (MOG35-55)-induced T-cell activation. We found that genetic loss of ADAM12 profoundly alleviated Th1-mediated neuroinflammation and thus disease severity in experimental autoimmune encephalomyelitis, a model of multiple sclerosis. Transcriptomic profiling of MOG35-55-specific ADAM12-/- T cells revealed differentially expressed genes that are important for T-cell activation, proliferation, and costimulatory signaling and Th1 pathogenicity, consistent with their inability to cause T-cell-mediated skin inflammation in a model of adoptive delayed-type hypersensitivity. We conclude that ADAM12 is a T-cell costimulatory molecule that contributes to the pathogenesis of tissue inflammation and a potential target for the treatment of Th1-mediated diseases.
Collapse
Affiliation(s)
- Yawei Liu
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Robert Bockermann
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Mahdieh Hadi
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Iman Safari
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Belinda Carrion
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Marie Kveiborg
- BRIC, Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
184
|
Saed B, Munaweera R, Anderson J, O'Neill WD, Hu YS. Rapid statistical discrimination of fluorescence images of T cell receptors on immobilizing surfaces with different coating conditions. Sci Rep 2021; 11:15488. [PMID: 34326382 PMCID: PMC8322097 DOI: 10.1038/s41598-021-94730-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/15/2021] [Indexed: 11/24/2022] Open
Abstract
The spatial organization of T cell receptors (TCRs) correlates with membrane-associated signal amplification, dispersion, and regulation during T cell activation. Despite its potential clinical importance, quantitative analysis of the spatial arrangement of TCRs from standard fluorescence images remains difficult. Here, we report Statistical Classification Analyses of Membrane Protein Images or SCAMPI as a technique capable of analyzing the spatial arrangement of TCRs on the plasma membrane of T cells. We leveraged medical image analysis techniques that utilize pixel-based values. We transformed grayscale pixel values from fluorescence images of TCRs into estimated model parameters of partial differential equations. The estimated model parameters enabled an accurate classification using linear discrimination techniques, including Fisher Linear Discriminant (FLD) and Logistic Regression (LR). In a proof-of-principle study, we modeled and discriminated images of fluorescently tagged TCRs from Jurkat T cells on uncoated cover glass surfaces (Null) or coated cover glass surfaces with either positively charged poly-L-lysine (PLL) or TCR cross-linking anti-CD3 antibodies (OKT3). Using 80 training images and 20 test images per class, our statistical technique achieved 85% discrimination accuracy for both OKT3 versus PLL and OKT3 versus Null conditions. The run time of image data download, model construction, and image discrimination was 21.89 s on a laptop computer, comprised of 20.43 s for image data download, 1.30 s on the FLD-SCAMPI analysis, and 0.16 s on the LR-SCAMPI analysis. SCAMPI represents an alternative approach to morphology-based qualifications for discriminating complex patterns of membrane proteins conditioned on a small sample size and fast runtime. The technique paves pathways to characterize various physiological and pathological conditions using the spatial organization of TCRs from patient T cells.
Collapse
Affiliation(s)
- Badeia Saed
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Rangika Munaweera
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Jesse Anderson
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - William D O'Neill
- Department of Bioengineering, Colleges of Engineering and Medicine, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| | - Ying S Hu
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
185
|
Kim B, Kim HY, Lee WW. Zap70 Regulates TCR-Mediated Zip6 Activation at the Immunological Synapse. Front Immunol 2021; 12:687367. [PMID: 34394081 PMCID: PMC8358678 DOI: 10.3389/fimmu.2021.687367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/15/2021] [Indexed: 01/23/2023] Open
Abstract
The essential microelement zinc plays immunoregulatory roles via its ability to influence signaling pathways. Zinc deficiency impairs overall immune function and resultantly increases susceptibility to infection. Thus, zinc is considered as an immune-boosting supplement for populations with hypozincemia at high-risk for infection. Besides its role as a structural cofactor of many proteins, zinc also acts as an intracellular messenger in immune cell signaling. T-cell activation instructs zinc influx from extracellular and subcellular sources through the Zip6 and Zip8 zinc transporters, respectively. Increased cytoplasmic zinc participates in the regulation of T-cell responses by modifying activation signaling. However, the mechanism underlying the activation-dependent movement of zinc ions by Zip transporters in T cells remains elusive. Here, we demonstrate that Zip6, one of the most abundantly expressed Zip transporters in T cells, is mainly localized to lipid rafts in human T cells and is recruited into the immunological synapse in response to TCR stimulation. This was demonstrated through confocal imaging of the interaction between CD4+ T cells and antigen-presenting cells. Further, immunoprecipitation assays show that TCR triggering induces tyrosine phosphorylation of Zip6, which has at least three putative tyrosine motifs in its long cytoplasmic region, and this phosphorylation is coupled with its physical interaction with Zap70. Silencing Zip6 reduces zinc influx from extracellular sources and suppresses T-cell responses, suggesting an interaction between Zip6-mediated zinc influx and TCR activation. These results provide new insights into the mechanism through which Zip6-mediated zinc influx occurs in a TCR activation-dependent manner in human CD4+ T cells.
Collapse
Affiliation(s)
- Bonah Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee Young Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea
| |
Collapse
|
186
|
Cassioli C, Balint S, Compeer EB, Felce JH, Gamberucci A, Della Bella C, Felce SL, Brunetti J, Valvo S, Pende D, D’Elios MM, Moretta L, Dustin ML, Baldari CT. Increasing LFA-1 Expression Enhances Immune Synapse Architecture and T Cell Receptor Signaling in Jurkat E6.1 Cells. Front Cell Dev Biol 2021; 9:673446. [PMID: 34368126 PMCID: PMC8343233 DOI: 10.3389/fcell.2021.673446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/21/2021] [Indexed: 11/21/2022] Open
Abstract
The Jurkat E6.1 clone has been extensively used as a powerful tool for the genetic and biochemical dissection of the TCR signaling pathway. More recently, these cells have been exploited in imaging studies to identify key players in immunological synapse (IS) assembly in superantigen-specific conjugates and to track the dynamics of signaling molecules on glass surfaces coated with activating anti-CD3 antibodies. By comparison, Jurkat cells have been used only scantily for imaging on supported lipid bilayers (SLBs) incorporating laterally mobile TCR and integrin ligands, which allow to study synaptic rearrangements of surface molecules and the fine architecture of the mature IS, likely due to limitations in the assembly of immune synapses with well-defined architecture. Here we have explored whether upregulating the low levels of endogenous LFA-1 expression on Jurkat E6.1 cells through transduction with CD11a- and CD18-encoding lentiviruses can improve IS architecture. We show that, while forced LFA-1 expression did not affect TCR recruitment to the IS, E6.1 LFA-1 high cells assembled better structured synapses, with a tighter distribution of signaling-competent TCRs at the center of the IS. LFA-1 upregulation enhanced protein phosphotyrosine signaling on SLBs but not at the IS formed in conjugates with SEE-pulsed APCs, and led to the constitutive formation of an intracellular phosphotyrosine pool co-localizing with endosomal CD3ζ. This was paralleled by an increase in the levels of p-ZAP-70 and p-Erk both under basal conditions and following activation, and in enhanced Ca2+ mobilization from intracellular stores. The enhancement in early signaling E6.1 LFA-1 high cells did not affect expression of the early activation marker CD69 but led to an increase in IL-2 expression. Our results highlight a new role for LFA-1 in the core architecture of the IS that can be exploited to study the spatiotemporal redistribution of surface receptors on SLBs, thereby extending the potential of E6.1 cells and their derivatives for fine-scale imaging studies.
Collapse
Affiliation(s)
- Chiara Cassioli
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Stefan Balint
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Ewoud B. Compeer
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - James H. Felce
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Chiara Della Bella
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Suet Ling Felce
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jlenia Brunetti
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Salvatore Valvo
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Daniela Pende
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Mario M. D’Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Michael L. Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
187
|
Ramachandran A, Mehić M, Wasim L, Malinova D, Gori I, Blaszczyk BK, Carvalho DM, Shore EM, Jones C, Hyvönen M, Tolar P, Hill CS. Pathogenic ACVR1 R206H activation by Activin A-induced receptor clustering and autophosphorylation. EMBO J 2021; 40:e106317. [PMID: 34003511 PMCID: PMC8280795 DOI: 10.15252/embj.2020106317] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 11/23/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) and diffuse intrinsic pontine glioma (DIPG) are debilitating diseases that share causal mutations in ACVR1, a TGF-β family type I receptor. ACVR1R206H is a frequent mutation in both diseases. Pathogenic signaling via the SMAD1/5 pathway is mediated by Activin A, but how the mutation triggers aberrant signaling is not known. We show that ACVR1 is essential for Activin A-mediated SMAD1/5 phosphorylation and is activated by two distinct mechanisms. Wild-type ACVR1 is activated by the Activin type I receptors, ACVR1B/C. In contrast, ACVR1R206H activation does not require upstream kinases, but is predominantly activated via Activin A-dependent receptor clustering, which induces its auto-activation. We use optogenetics and live-imaging approaches to demonstrate Activin A-induced receptor clustering and show it requires the type II receptors ACVR2A/B. Our data provide molecular mechanistic insight into the pathogenesis of FOP and DIPG by linking the causal activating genetic mutation to disrupted signaling.
Collapse
Affiliation(s)
- Anassuya Ramachandran
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | - Merima Mehić
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| | - Laabiah Wasim
- Immune Receptor Activation LaboratoryThe Francis Crick InstituteLondonUK
| | | | - Ilaria Gori
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| | | | - Diana M Carvalho
- Division of Molecular PathologyThe Institute of Cancer ResearchSuttonUK
| | - Eileen M Shore
- Departments of Orthopaedic Surgery and GeneticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Chris Jones
- Division of Molecular PathologyThe Institute of Cancer ResearchSuttonUK
| | - Marko Hyvönen
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Pavel Tolar
- Immune Receptor Activation LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
Division of Infection and ImmunityInstitute of Immunity and TransplantationUniversity CollegeLondonUK
| | - Caroline S Hill
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
188
|
Abstract
T cell activation is a critical event in the adaptive immune response, indispensable for cell-mediated and humoral immunity as well as for immune regulation. Recent years have witnessed an emerging trend emphasizing the essential role that physical force and mechanical properties play at the T cell interface. In this review, we integrate current knowledge of T cell antigen recognition and the different models of T cell activation from the perspective of mechanobiology, focusing on the interaction between the T cell receptor (TCR) and the peptide-major histocompatibility complex (pMHC) antigen. We address the shortcomings of TCR affinity alone in explaining T cell functional outcomes and the rising status of force-regulated TCR bond lifetimes, most notably the TCR catch bond. Ultimately, T cell activation and the ensuing physiological responses result from mechanical interaction between TCRs and the pMHC. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Baoyu Liu
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| | - Elizabeth M Kolawole
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| | - Brian D Evavold
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| |
Collapse
|
189
|
German Y, Vulliard L, Kamnev A, Pfajfer L, Huemer J, Mautner AK, Rubio A, Kalinichenko A, Boztug K, Ferrand A, Menche J, Dupré L. Morphological profiling of human T and NK lymphocytes by high-content cell imaging. Cell Rep 2021; 36:109318. [PMID: 34233185 DOI: 10.1016/j.celrep.2021.109318] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 02/25/2021] [Accepted: 06/07/2021] [Indexed: 01/21/2023] Open
Abstract
The immunological synapse is a complex structure that decodes stimulatory signals into adapted lymphocyte responses. It is a unique window to monitor lymphocyte activity because of development of systematic quantitative approaches. Here we demonstrate the applicability of high-content imaging to human T and natural killer (NK) cells and develop a pipeline for unbiased analysis of high-definition morphological profiles. Our approach reveals how distinct facets of actin cytoskeleton remodeling shape immunological synapse architecture and affect lytic granule positioning. Morphological profiling of CD8+ T cells from immunodeficient individuals allows discrimination of the roles of the ARP2/3 subunit ARPC1B and the ARP2/3 activator Wiskott-Aldrich syndrome protein (WASP) in immunological synapse assembly. Single-cell analysis further identifies uncoupling of lytic granules and F-actin radial distribution in ARPC1B-deficient lymphocytes. Our study provides a foundation for development of morphological profiling as a scalable approach to monitor primary lymphocyte responsiveness and to identify complex aspects of lymphocyte micro-architecture.
Collapse
Affiliation(s)
- Yolla German
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM UMR1291, CNRS UMR5051, Toulouse III Paul Sabatier University, Toulouse, France; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria
| | - Loan Vulliard
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Vienna, Austria
| | - Anton Kamnev
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Laurène Pfajfer
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM UMR1291, CNRS UMR5051, Toulouse III Paul Sabatier University, Toulouse, France; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria
| | - Jakob Huemer
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria; St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Anna-Katharina Mautner
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Aude Rubio
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, 31024 Toulouse, France
| | - Artem Kalinichenko
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria; St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria; St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Audrey Ferrand
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, 31024 Toulouse, France
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Vienna, Austria; Faculty of Mathematics, University of Vienna, Vienna, Austria
| | - Loïc Dupré
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM UMR1291, CNRS UMR5051, Toulouse III Paul Sabatier University, Toulouse, France; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
190
|
Dupré L, Boztug K, Pfajfer L. Actin Dynamics at the T Cell Synapse as Revealed by Immune-Related Actinopathies. Front Cell Dev Biol 2021; 9:665519. [PMID: 34249918 PMCID: PMC8266300 DOI: 10.3389/fcell.2021.665519] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/06/2021] [Indexed: 01/21/2023] Open
Abstract
The actin cytoskeleton is composed of dynamic filament networks that build adaptable local architectures to sustain nearly all cellular activities in response to a myriad of stimuli. Although the function of numerous players that tune actin remodeling is known, the coordinated molecular orchestration of the actin cytoskeleton to guide cellular decisions is still ill defined. T lymphocytes provide a prototypical example of how a complex program of actin cytoskeleton remodeling sustains the spatio-temporal control of key cellular activities, namely antigen scanning and sensing, as well as polarized delivery of effector molecules, via the immunological synapse. We here review the unique knowledge on actin dynamics at the T lymphocyte synapse gained through the study of primary immunodeficiences caused by mutations in genes encoding actin regulatory proteins. Beyond the specific roles of individual actin remodelers, we further develop the view that these operate in a coordinated manner and are an integral part of multiple signaling pathways in T lymphocytes.
Collapse
Affiliation(s)
- Loïc Dupré
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Laurène Pfajfer
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France.,St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| |
Collapse
|
191
|
Strategies for monitoring cell-cell interactions. Nat Chem Biol 2021; 17:641-652. [PMID: 34035514 DOI: 10.1038/s41589-021-00790-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/30/2021] [Indexed: 02/03/2023]
Abstract
Multicellular organisms depend on physical cell-cell interactions to control physiological processes such as tissue formation, neurotransmission and immune response. These intercellular binding events can be both highly dynamic in their duration and complex in their composition, involving the participation of many different surface and intracellular biomolecules. Untangling the intricacy of these interactions and the signaling pathways they modulate has greatly improved insight into the biological processes that ensue upon cell-cell engagement and has led to the development of protein- and cell-based therapeutics. The importance of monitoring physical cell-cell interactions has inspired the development of several emerging approaches that effectively interrogate cell-cell interfaces with molecular-level detail. Specifically, the merging of chemistry- and biology-based technologies to deconstruct the complexity of cell-cell interactions has provided new avenues for understanding cell-cell interaction biology and opened opportunities for therapeutic development.
Collapse
|
192
|
Gilfillan CB, Hebeisen M, Rufer N, Speiser DE. Constant regulation for stable CD8 T-cell functional avidity and its possible implications for cancer immunotherapy. Eur J Immunol 2021; 51:1348-1360. [PMID: 33704770 PMCID: PMC8252569 DOI: 10.1002/eji.202049016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/16/2020] [Accepted: 03/05/2021] [Indexed: 12/30/2022]
Abstract
The functional avidity (FA) of cytotoxic CD8 T cells impacts strongly on their functional capabilities and correlates with protection from infection and cancer. FA depends on TCR affinity, downstream signaling strength, and TCR affinity-independent parameters of the immune synapse, such as costimulatory and inhibitory receptors. The functional impact of coreceptors on FA remains to be fully elucidated. Despite its importance, FA is infrequently assessed and incompletely understood. There is currently no consensus as to whether FA can be enhanced by optimized vaccine dose or boosting schedule. Recent findings suggest that FA is remarkably stable in vivo, possibly due to continued signaling modulation of critical receptors in the immune synapse. In this review, we provide an overview of the current knowledge and hypothesize that in vivo, codominant T cells constantly "equalize" their FA for similar function. We present a new model of constant FA regulation, and discuss practical implications for T-cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Connie B. Gilfillan
- Department of OncologyUniversity Hospital and University of LausanneLausanneSwitzerland
| | - Michael Hebeisen
- Department of OncologyUniversity Hospital and University of LausanneLausanneSwitzerland
| | - Nathalie Rufer
- Department of OncologyUniversity Hospital and University of LausanneLausanneSwitzerland
| | - Daniel E. Speiser
- Department of OncologyUniversity Hospital and University of LausanneLausanneSwitzerland
| |
Collapse
|
193
|
Bhingardive V, Kossover A, Iraqi M, Khand B, Le Saux G, Porgador A, Schvartzman M. Antibody-Functionalized Nanowires: A Tuner for the Activation of T Cells. NANO LETTERS 2021; 21:4241-4248. [PMID: 33989498 DOI: 10.1021/acs.nanolett.1c00245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
T cells sense both chemical cues delivered by antigen molecules and physical cues delivered by the environmental elasticity and topography; yet, it is still largely unclear how these cues cumulatively regulate the immune activity of T cells. Here, we engineered a nanoscale platform for ex vivo stimulation of T cells based on antigen-functionalized nanowires. The nanowire topography and elasticity, as well as the immobilized antigens, deliver the physical and chemical cues, respectively, enabling the systematic study of the integrated effect of these cues on a T cell's immune response. We found that T cells sense both the topography and bending modulus of the nanowires and modulate their signaling, degranulation, and cytotoxicity with the variation in these physical features. Our study provides an important insight into the physical mechanism of T cell activation and paves the way to novel nanomaterials for the controlled ex vivo activation of T cells in immunotherapy.
Collapse
|
194
|
Verron Q, Forslund E, Brandt L, Leino M, Frisk TW, Olofsson PE, Önfelt B. NK cells integrate signals over large areas when building immune synapses but require local stimuli for degranulation. Sci Signal 2021; 14:14/684/eabe2740. [PMID: 34035142 DOI: 10.1126/scisignal.abe2740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Immune synapses are large-scale, transient molecular assemblies that serve as platforms for antigen presentation to B and T cells and for target recognition by cytotoxic T cells and natural killer (NK) cells. The formation of an immune synapse is a tightly regulated, stepwise process in which the cytoskeleton, cell surface receptors, and intracellular signaling proteins rearrange into supramolecular activation clusters (SMACs). We generated artificial immune synapses (AIS) consisting of synthetic and natural ligands for the NK cell-activating receptors LFA-1 and CD16 by microcontact printing the ligands into circular-shaped SMAC structures. Live-cell imaging and analysis of fixed human NK cells in this reductionist system showed that the spatial distribution of activating ligands influenced the formation, stability, and outcome of NK cell synapses. Whereas engagement of LFA-1 alone promoted synapse initiation, combined engagement of LFA-1 and CD16 was required for the formation of mature synapses and degranulation. Organizing LFA-1 and CD16 ligands into donut-shaped AIS resulted in fewer long-lasting, symmetrical synapses compared to dot-shaped AIS. NK cells spreading evenly over either AIS shape exhibited similar arrangements of the lytic machinery. However, degranulation only occurred in regions containing ligands that therefore induced local signaling, suggesting the existence of a late checkpoint for degranulation. Our results demonstrate that the spatial organization of ligands in the synapse can affect its outcome, which could be exploited by target cells as an escape mechanism.
Collapse
Affiliation(s)
- Quentin Verron
- Biophysics, Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Elin Forslund
- Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Ludwig Brandt
- Biophysics, Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mattias Leino
- Biophysics, Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Thomas W Frisk
- Biophysics, Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Per E Olofsson
- Biophysics, Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Björn Önfelt
- Biophysics, Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden. .,Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
195
|
WASP and Mst1 coregulate B-cell development and B-cell receptor signaling. Blood Adv 2021; 4:573-585. [PMID: 32045478 DOI: 10.1182/bloodadvances.2018027870] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/11/2019] [Indexed: 12/25/2022] Open
Abstract
Mst1 is a serine/threonine kinase involved in cell survival, proliferation, apoptosis, and tumorigenesis. In mice, Mst1 regulates actin dynamics required for T-cell adhesion and migration, which correlate with thymic egress and entry into lymphatic tissue. The role of Mst1 in B cells and how it may control actin-dependent processes has not been well characterized. Wiskott-Aldrich syndrome protein (WASP) deficiency only moderately affects development and B-cell receptor (BCR) signaling, suggesting WASP likely associates with other molecules. We investigated whether Mst1 associates with WASP to regulate B-cell development and activation. Experimenting on Mst1/WASP double knockout (DKO) mice, we found a severe defect in the bone marrow B-cell development, and BCR signaling in the DKO mice was severely reduced. Even though WASP or Mst1 could influence the early B-cell activation, we found that the early activation events such as B-cell spreading, BCR clustering, and BCR signaling were much more impaired in the B cells from DKO mice. Furthermore, reciprocal regulation between Mst1 and WASP was observed in WASP and Mst1 KO mice, whereby the localization and function of phosphorylated WASP were affected in Mst1 KO mice. Most importantly, Mst1 inhibits the expression of WASP by decreasing the expression of WASP-interacting protein. Interestingly, we also found that WASP deficiency in patients and mice interferes with phosphorylated Mst1 localization and therefore function in B cells. Overall, our study provides a partner for WASP to regulate B-cell development and BCR signaling, as well as the reciprocal regulating molecular mechanism of one another.
Collapse
|
196
|
Takehara T, Wakamatsu E, Machiyama H, Nishi W, Emoto K, Azuma M, Soejima K, Fukunaga K, Yokosuka T. PD-L2 suppresses T cell signaling via coinhibitory microcluster formation and SHP2 phosphatase recruitment. Commun Biol 2021; 4:581. [PMID: 33990697 PMCID: PMC8121797 DOI: 10.1038/s42003-021-02111-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/14/2021] [Indexed: 01/08/2023] Open
Abstract
The coinhibitory receptor, PD-1, is of major importance for the suppression of T cell activation in various types of immune responses. A high-resolution imaging study showed that PD-1 forms a coinhibitory signalosome, "PD-1 microcluster", with the phosphatase, SHP2, to dephosphorylate the TCR/CD3 complex and its downstream signaling molecules. Such a consecutive reaction entirely depended on PD-1-PD-L1/2 binding. PD-L2 is expressed on professional antigen-presenting cells and also on some tumor cells, which possibly explains the discrepant efficacy of immune checkpoint therapy for PD-L1-negative tumors. Here, we performed precise imaging analysis of PD-L2 forming PD-1-PD-L2 clusters associating with SHP2. PD-L2 could compete with PD-L1 for binding to PD-1, occupying the same space at TCR microclusters. The PD-1 microcluster formation was inhibited by certain mAbs with functional consequences. Thus, PD-1 microcluster formation provides a visible index for the effectiveness of anti-PD-1- or anti-PD-L1/2-mediated T cell suppression. PD-L2 may exert immune suppressive responses cooperatively with PD-L1 on the microcluster scale.
Collapse
Affiliation(s)
- Tomohiro Takehara
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Immunology, Tokyo Medical University, Tokyo, Japan
| | - Ei Wakamatsu
- Department of Immunology, Tokyo Medical University, Tokyo, Japan
| | | | - Wataru Nishi
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsura Emoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenzo Soejima
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tadashi Yokosuka
- Department of Immunology, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
197
|
Göhring J, Kellner F, Schrangl L, Platzer R, Klotzsch E, Stockinger H, Huppa JB, Schütz GJ. Temporal analysis of T-cell receptor-imposed forces via quantitative single molecule FRET measurements. Nat Commun 2021; 12:2502. [PMID: 33947864 PMCID: PMC8096839 DOI: 10.1038/s41467-021-22775-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 03/25/2021] [Indexed: 02/01/2023] Open
Abstract
Mechanical forces acting on ligand-engaged T-cell receptors (TCRs) have previously been implicated in T-cell antigen recognition, yet their magnitude, spread, and temporal behavior are still poorly defined. We here report a FRET-based sensor equipped either with a TCR-reactive single chain antibody fragment or peptide-loaded MHC, the physiological TCR-ligand. The sensor was tethered to planar glass-supported lipid bilayers (SLBs) and informed most directly on the magnitude and kinetics of TCR-imposed forces at the single molecule level. When confronting T-cells with gel-phase SLBs we observed both prior and upon T-cell activation a single, well-resolvable force-peak of approximately 5 pN and force loading rates on the TCR of 1.5 pN per second. When facing fluid-phase SLBs instead, T-cells still exerted tensile forces yet of threefold reduced magnitude and only prior to but not upon activation.
Collapse
Affiliation(s)
- Janett Göhring
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Institute of Applied Physics, TU Wien, Vienna, Austria
| | - Florian Kellner
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - René Platzer
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Enrico Klotzsch
- Institute of Applied Physics, TU Wien, Vienna, Austria
- Laboratory of Applied Mechanobiology, Department for Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Institute of Biology, Experimental Biophysics/ Mechanobiology, Humboldt Universität zu Berlin, Berlin, Germany
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes B Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | | |
Collapse
|
198
|
Tian Z, Liu M, Zhang Y, Wang X. Bispecific T cell engagers: an emerging therapy for management of hematologic malignancies. J Hematol Oncol 2021; 14:75. [PMID: 33941237 PMCID: PMC8091790 DOI: 10.1186/s13045-021-01084-4] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Harnessing the power of immune cells, especially T cells, to enhance anti-tumor activities has become a promising strategy in clinical management of hematologic malignancies. The emerging bispecific antibodies (BsAbs), which recruit T cells to tumor cells, exemplified by bispecific T cell engagers (BiTEs), have facilitated the development of tumor immunotherapy. Here we discussed the advances and challenges in BiTE therapy developed for the treatment of hematologic malignancies. Blinatumomab, the first BiTE approved for the treatment of acute lymphocytic leukemia (ALL), is appreciated for its high efficacy and safety. Recent studies have focused on improving the efficacy of BiTEs by optimizing treatment regimens and refining the molecular structures of BiTEs. A considerable number of bispecific T cell-recruiting antibodies which are potentially effective in hematologic malignancies have been derived from BiTEs. The elucidation of mechanisms of BiTE action and neonatal techniques used for the construction of BsAbs can improve the treatment of hematological malignancies. This review summarized the features of bispecific T cell-recruiting antibodies for the treatment of hematologic malignancies with special focus on preclinical experiments and clinical studies.
Collapse
Affiliation(s)
- Zheng Tian
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Ming Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China.,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China. .,School of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China. .,School of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
199
|
Grochmal A, Woods B, Milanesi L, Perez-Soto M, Tomas S. How the biomimetic assembly of membrane receptors into multivalent domains is regulated by a small ligand. Chem Sci 2021; 12:7800-7808. [PMID: 34168834 PMCID: PMC8188472 DOI: 10.1039/d1sc01598b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/30/2021] [Indexed: 11/21/2022] Open
Abstract
In living cells, communication requires the action of membrane receptors that are activated following very small environmental changes. A binary all-or-nothing behavior follows, making the organism extremely efficient at responding to specific stimuli. Using a minimal system composed of lipid vesicles, chemical models of a membrane receptor and their ligands, we show that bio-mimetic ON/OFF assembly of high avidity, multivalent domains is triggered by small temperature changes. Moreover, the intensity of the ON signal at the onset of the switch is modulated by the presence of small, weakly binding divalent ligands, reminiscent of the action of primary messengers in biological systems. Based on the analysis of spectroscopic data, we develop a mathematical model that rigorously describes the temperature-dependent switching of the membrane receptor assembly and ligand binding. From this we derive an equation that predicts the intensity of the modulation of the ON signal by the ligand-messenger as a function of the pairwise binding parameters, the number of binding sites that it features and the concentration. The behavior of our system, and the model derived, highlight the usefulness of weakly binding ligands in the regulation of membrane receptors and the pitfalls inherent to their binding promiscuity, such as non-specific binding to the membrane. Our model, and the equations derived from it, offer a valuable tool for the study of membrane receptors in both biological and biomimetic settings. The latter can be exploited to program membrane receptor avidity on sensing vesicles, create hierarchical protocell tissues or develop highly specific drug delivery vehicles.
Collapse
Affiliation(s)
- Anna Grochmal
- Department of Biological Sciences, Institute of Structural and Molecular Biology, School of Science, Birkbeck University of London Malet Street London WC1E 7HX UK
| | - Ben Woods
- Department of Biological Sciences, Institute of Structural and Molecular Biology, School of Science, Birkbeck University of London Malet Street London WC1E 7HX UK
| | - Lilia Milanesi
- Department of Biological Sciences, Institute of Structural and Molecular Biology, School of Science, Birkbeck University of London Malet Street London WC1E 7HX UK
| | - Manuel Perez-Soto
- Department of Biological Sciences, Institute of Structural and Molecular Biology, School of Science, Birkbeck University of London Malet Street London WC1E 7HX UK
| | - Salvador Tomas
- Department of Biological Sciences, Institute of Structural and Molecular Biology, School of Science, Birkbeck University of London Malet Street London WC1E 7HX UK
| |
Collapse
|
200
|
Jha V, Nicholson LK, Gardner EM, Rahkola JT, Pratap H, Scott J, Borgeson M, Jacobelli J, Janoff EN. Impact of HIV-1 Infection and Antigen Class on T Follicular Helper Cell Responses to Pneumococcal Polysaccharide-Protein Conjugate Vaccine-13. THE JOURNAL OF IMMUNOLOGY 2021; 206:2402-2411. [PMID: 33931485 DOI: 10.4049/jimmunol.2001133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/04/2021] [Indexed: 11/19/2022]
Abstract
Pneumococcal infections are common and serious complications of HIV-1 disease. Prevention has been compromised by the limited magnitude and quality of Ab responses to T cell-independent type 2 pneumococcal capsular polysaccharides (PPS). The pneumococcal polysaccharide-protein conjugate vaccine-13 (PCV-13) contains PPS conjugated to the T cell-dependent protein (diphtheria toxoid [DT] [CRM197]). We investigated the differential response to PPS and DT by human Ab-secreting B cells (ASC) after immunization with PCV-13 in newly diagnosed healthy HIV+ and control adults. The numbers of PPS-specific IgG ASC increased significantly and similarly in HIV+ and controls. However, DT-specific IgG ASC increased in controls but not HIV+ subjects. To determine the cellular basis of these disparate responses to DT and PPS, we characterized the frequency and activation of T follicular helper (Tfh) cells, the predominant T cell subset providing B cell help. Expression of inducible T cell costimulator (ICOS), which sustains Tfh function and phenotype, increased significantly among controls, when compared with the HIV+ group. Increases in ICOS+ Tfh correlated with changes in T-dependent, DT-specific IgG ASC in controls but not in HIV+ In contrast, ICOS expression did not correlate with T cell-independent type 2 PPS-specific ASC in either group. Of note, upon optimized ex vivo stimulation, CD4 T cells from HIV+ subjects differentiated into Tfh cells and formed synapses with Raji B cells at frequencies similar to that of controls. In summary, PCV-13-induced increase in ICOS expression on Tfh was associated with responses to DT, which was compromised in recently diagnosed healthy HIV+ adults and can be restored ex vivo by providing effective Tfh-differentiating signals.
Collapse
Affiliation(s)
- Vibha Jha
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Lindsay K Nicholson
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | | | - Jeremy T Rahkola
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Barbara Davis Center, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Harsh Pratap
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | | | - Mandy Borgeson
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Jordan Jacobelli
- Barbara Davis Center, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Edward N Janoff
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO .,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|