51
|
Hickey JW, Dong Y, Chung JW, Salathe SF, Pruitt HC, Li X, Chang C, Fraser AK, Bessell CA, Ewald AJ, Gerecht S, Mao HQ, Schneck JP. Engineering an Artificial T-Cell Stimulating Matrix for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807359. [PMID: 30968468 PMCID: PMC8601018 DOI: 10.1002/adma.201807359] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/04/2019] [Indexed: 05/17/2023]
Abstract
T cell therapies require the removal and culture of T cells ex vivo to expand several thousand-fold. However, these cells often lose the phenotype and cytotoxic functionality for mediating effective therapeutic responses. The extracellular matrix (ECM) has been used to preserve and augment cell phenotype; however, it has not been applied to cellular immunotherapies. Here, a hyaluronic acid (HA)-based hydrogel is engineered to present the two stimulatory signals required for T-cell activation-termed an artificial T-cell stimulating matrix (aTM). It is found that biophysical properties of the aTM-stimulatory ligand density, stiffness, and ECM proteins-potentiate T cell signaling and skew phenotype of both murine and human T cells. Importantly, the combination of the ECM environment and mechanically sensitive TCR signaling from the aTM results in a rapid and robust expansion of rare, antigen-specific CD8+ T cells. Adoptive transfer of these tumor-specific cells significantly suppresses tumor growth and improves animal survival compared with T cells stimulated by traditional methods. Beyond immediate immunotherapeutic applications, demonstrating the environment influences the cellular therapeutic product delineates the importance of the ECM and provides a case study of how to engineer ECM-mimetic materials for therapeutic immune stimulation in the future.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Institute for Cell Engineering, School of Medicine, Baltimore, MD, 21205, USA
- Department of Pathology, School of Medicine, Baltimore, MD, 21287, USA
- Translational Tissue Engineering Center, Baltimore, MD, 21287, USA
- Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
| | - Yi Dong
- Graduate Program in Immunology, School of Medicine, Baltimore, MD, 21205, USA
| | - Jae Wook Chung
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
| | - Sebastian F Salathe
- Department of Biology, Krieger School of Arts and Sciences, Baltimore, MD, 21218, USA
| | - Hawley C Pruitt
- Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
| | - Xiaowei Li
- Translational Tissue Engineering Center, Baltimore, MD, 21287, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
| | - Calvin Chang
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Baltimore, MD, 21287, USA
| | - Andrew K Fraser
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Baltimore, MD, 21205, USA
| | - Catherine A Bessell
- Graduate Program in Immunology, School of Medicine, Baltimore, MD, 21205, USA
| | - Andrew J Ewald
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Baltimore, MD, 21205, USA
- Department of Oncology, School of Medicine, Baltimore, MD, 21205, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
- Physical Sciences-Oncology Center, Baltimore, MD, 21218, USA
| | - Hai-Quan Mao
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Baltimore, MD, 21287, USA
- Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
| | - Jonathan P Schneck
- Institute for Cell Engineering, School of Medicine, Baltimore, MD, 21205, USA
- Department of Pathology, School of Medicine, Baltimore, MD, 21287, USA
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| |
Collapse
|
52
|
Torralba D, Martín-Cófreces NB, Sanchez-Madrid F. Mechanisms of polarized cell-cell communication of T lymphocytes. Immunol Lett 2019; 209:11-20. [PMID: 30954509 DOI: 10.1016/j.imlet.2019.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/13/2019] [Accepted: 03/17/2019] [Indexed: 01/07/2023]
Abstract
Cell-cell communication comprises a variety of molecular mechanisms that immune cells use to respond appropriately to diverse pathogenic stimuli. T lymphocytes polarize in response to different stimuli, such as cytokines, adhesion to specific ligands and cognate antigens presented in the context of MHC. Polarization takes different shapes, from migratory front-back polarization to the formation of immune synapses (IS). The formation of IS between a T cell and an antigen-presenting cell involves early events of receptor-ligand interaction leading to the reorganization of the plasma membrane and the cytoskeleton to orchestrate vesicular and endosomal traffic and directed secretion of several types of mediators, including cytokines and nanovesicles. Cell polarization involves the repositioning of many subcellular organelles, including the endosomal compartment, which becomes an effective platform for the shuttling of molecules as vesicular cargoes that lately will be secreted to transfer information to antigen-presenting cells. Overall, the polarized interaction between a T cell and APC modifies the recipient cell in different ways that are likely lineage-dependent, e.g. dendritic cells, B cells or even other T cells. In this review, we will discuss the mechanisms that mediate the polarization of different membrane receptors, cytoskeletal components and organelles in T cells in a variety of immune contexts.
Collapse
Affiliation(s)
- D Torralba
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, 28006 Madrid, Spain; Area of Vascular Pathophysiology, Laboratory of Intercellular Communication Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - N B Martín-Cófreces
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, 28006 Madrid, Spain; Area of Vascular Pathophysiology, Laboratory of Intercellular Communication Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - F Sanchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, 28006 Madrid, Spain; Area of Vascular Pathophysiology, Laboratory of Intercellular Communication Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| |
Collapse
|
53
|
Abstract
T cells effectively explore the tissue in search for antigens. When activated, they dedicate a big amount of energy and resources to arrange a complex structure called immunological synapse (IS), containing a particular distribution of molecules defined as supramolecular activation clusters (SMACs), and become polarized toward the target cell in a manner that channels the information specifically. This arrangement is symmetrical and requires the polarization of the MTOC and the Golgi to be operational, especially for the proper delivery of lytic granules and the recycling of molecules three dimensionally segregated at the clustered interface. Alternatively, after the productive encounter, T cells need to rearrange again to newly navigate through the tissue, changing back to a motile state called immunological kinapse (IK). In this IK state, the MTOC and the Golgi apparatus are repositioned and recruited at the back of the T cell to facilitate motility, while the established symmetry of the elements of the SMACs is broken and distributed in a different pattern. Both states, IS and IK, are interchangeable and are mainly orchestrated by the MTOC/Golgi complex, being critical for an effective immune response.
Collapse
|
54
|
Cai G, Sun K, Wang T, Zou H, Gu J, Yuan Y, Liu X, Liu Z, Bian J. Mechanism and effects of Zearalenone on mouse T lymphocytes activation in vitro. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 162:208-217. [PMID: 29990733 DOI: 10.1016/j.ecoenv.2018.06.055] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/16/2018] [Accepted: 06/19/2018] [Indexed: 06/08/2023]
Abstract
Zearalenone (ZEA) is particularly toxic to the female reproductive system. Nevertheless, the effect of ZEA on the immune system is still not fully understood. The following study investigates the effects and mechanism of ZEA on mouse T cell activation in vitro. Briefly, T lymphocytes were extracted from primary splenic lymphocyte in mice, activated by concanavalin A, and then were exposed to different concentrations of ZEA for a certain period of time. Flow cytometry was used to detect the expression of activating and co-stimulatory molecules, and the secretion of cytokines in T cells at various stages. The expression of initiation regulatory protein in T cell activation, nuclear factor protein and co-stimulatory molecule related PI3K-Akt-mTOR signaling pathway proteins were detected by western blot. Our data showed that ZEA exposure inhibits the activity of T cell, and inhibits the expression of different activation signals in T cell. Additionally, ZEA exposure reduces the expression of initiative regulatory protein, i.e. LAT, Lck, Zap-70 during the activation of T cells. Thus, the results showed that ZEA exposure inhibits the formation and transmission of activated signal in T cells, interferes with signal pathway of T cell activation nuclear factor NFAT and NFκB, and decreases the secretion of cytokines after activation. Moreover, ZEA exposure interferes with co-stimulatory molecule CD28 during T cell activation, and with the activity of the PI3K-Akt-mTOR signaling pathway downstream of CD28. To conclude, our results indicated that ZEA toxin interferes with the activation of mouse T lymphocytes by affecting TCR signal and co-stimulatory signal, thus playing an essential role in immune toxicity.
Collapse
Affiliation(s)
- Guodong Cai
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Kai Sun
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Xuezhong Liu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China.
| |
Collapse
|
55
|
Kwun J, Park J, Yi JS, Farris AB, Kirk AD, Knechtle SJ. IL-21 Biased Alemtuzumab Induced Chronic Antibody-Mediated Rejection Is Reversed by LFA-1 Costimulation Blockade. Front Immunol 2018; 9:2323. [PMID: 30374350 PMCID: PMC6196291 DOI: 10.3389/fimmu.2018.02323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/18/2018] [Indexed: 11/25/2022] Open
Abstract
Despite its excellent efficacy in controlling T cell mediated acute rejection, lymphocyte depletion may promote a humoral response. While T cell repopulation after depletion has been evaluated in many aspects, the B cell response has not been fully elucidated. We tested the hypothesis that the mechanisms also involve skewed T helper phenotype after lymphocytic depletion. Post-transplant immune response was measured from alemtuzumab treated hCD52Tg cardiac allograft recipients with or without anti-LFA-1 mAb. Alemtuzumab induction promoted serum DSA, allo-B cells, and CAV in humanized CD52 transgenic (hCD52Tg) mice after heterotopic heart transplantation. Additional anti-LFA-1 mAb treatment resulted in reduced DSA (Fold increase 4.75 ± 6.9 vs. 0.7 ± 0.5; p < 0.01), allo-specific B cells (0.07 ± 0.06 vs. 0.006 ± 0.002 %; p < 0.01), neo-intimal hyperplasia (56 ± 14% vs. 23 ± 13%; p < 0.05), arterial disease (77.8 ± 14.2 vs. 25.8 ± 20.1%; p < 0.05), and fibrosis (15 ± 23.3 vs. 4.3 ± 1.65%; p < 0.05) in this alemtuzumab-induced chronic antibody-mediated rejection (CAMR) model. Surprisingly, elevated serum IL-21 levels in alemtuzumab-treated mice was reduced with LFA-1 blockade. In accordance with the increased serum IL-21 level, alemtuzumab treated mice showed hyperplastic germinal center (GC) development, while the supplemental anti-LFA-1 mAb significantly reduced the GC frequency and size. We report that the incomplete T cell depletion inside of the GC leads to a systemic IL-21 dominant milieu with hyperplastic GC formation and CAMR. Conventional immunosuppression, such as tacrolimus and rapamycin, failed to reverse AMR, while co-stimulation blockade with LFA-1 corrected the GC hyperplastic response. The identification of IL-21 driven chronic AMR elucidates a novel mechanism that suggests a therapeutic approach with cytolytic induction.
Collapse
Affiliation(s)
- Jean Kwun
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Jaeberm Park
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - John S Yi
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Alton B Farris
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
| | - Allan D Kirk
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Stuart J Knechtle
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
56
|
Martin-Blanco N, Blanco R, Alda-Catalinas C, Bovolenta ER, Oeste CL, Palmer E, Schamel WW, Lythe G, Molina-París C, Castro M, Alarcon B. A window of opportunity for cooperativity in the T Cell Receptor. Nat Commun 2018; 9:2618. [PMID: 29976994 PMCID: PMC6033938 DOI: 10.1038/s41467-018-05050-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 06/10/2018] [Indexed: 01/15/2023] Open
Abstract
The T-cell antigen receptor (TCR) is pre-organised in oligomers, known as nanoclusters. Nanoclusters could provide a framework for inter-TCR cooperativity upon peptide antigen-major histocompatibility complex (pMHC) binding. Here we have used soluble pMHC oligomers in search for cooperativity effects along the plasma membrane plane. We find that initial binding events favour subsequent pMHC binding to additional TCRs, during a narrow temporal window. This behaviour can be explained by a 3-state model of TCR transition from Resting to Active, to a final Inhibited state. By disrupting nanoclusters and hampering the Active conformation, we show that TCR cooperativity is consistent with TCR nanoclusters adopting the Active state in a coordinated manner. Preferential binding of pMHC to the Active TCR at the immunological synapse suggests that there is a transient time frame for signal amplification in the TCR, allowing the T cells to keep track of antigen quantity and binding time. T cells can be activated by a small, two-digit, number of antigen peptide molecules even though the receptor for antigen (TCR) is of low affinity. Here the authors present evidence that all TCRs within a nanocluster can become activated when only a subset is bound to antigen.
Collapse
Affiliation(s)
- N Martin-Blanco
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - R Blanco
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - C Alda-Catalinas
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - E R Bovolenta
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - C L Oeste
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - E Palmer
- University Hospital Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - W W Schamel
- Faculty of Biology, Institute Biology III, University of Freiburg, 79104, Freiburg, Germany.,Centre for Biological Signalling Studies (BIOSS), University of Freiburg, 79104, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center Freiburg and Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - G Lythe
- School of Mathematics, University of Leeds, Leeds, LS2 9JT, UK
| | - C Molina-París
- School of Mathematics, University of Leeds, Leeds, LS2 9JT, UK.
| | - M Castro
- School of Mathematics, University of Leeds, Leeds, LS2 9JT, UK. .,Grupo Interdisciplinar de Sistemas Complejos (GISC), Universidad Pontificia Comillas, Alberto Aguilera25, 28015, Madrid, Spain.
| | - B Alarcon
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049, Madrid, Spain.
| |
Collapse
|
57
|
Tai Y, Wang Q, Korner H, Zhang L, Wei W. Molecular Mechanisms of T Cells Activation by Dendritic Cells in Autoimmune Diseases. Front Pharmacol 2018; 9:642. [PMID: 29997500 PMCID: PMC6028573 DOI: 10.3389/fphar.2018.00642] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/29/2018] [Indexed: 12/21/2022] Open
Abstract
The interaction between T cell and dendritic cells (DCs) that leads to T cell activation affects the progression of the immune response including autoimmune diseases. Antigen presentation on immune cell surface, formation of an immunological synapse (IS), and specific identification of complex by T cells including two activating signals are necessary steps that lead to T cell activation. The formation of stimulatory IS involves the inclusion of costimulatory molecules, such as ICAM-1/LFA-1 and CD28/B7-1, and so on. Some fusion proteins and monoclonal antibodies targeting costimulatory molecules have been developed and approved to treat autoimmune diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), type I diabetes (T1D), inflammatory bowel disease (IBD), and psoriasis. These biological agents, including CTLA-4- and LFA-3-Ig, anti-CD3 monoclonal antibody, could prevent the successful engagement of DCs by T cell with significant efficacy and safety profile. In this article, we reviewed the molecular mechanisms of T cell activation during the interaction between T cells and DCs, and summarized some biological agents that target costimulatory molecules involved in the regulation of T cell activation.
Collapse
Affiliation(s)
- Yu Tai
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Qingtong Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Heinrich Korner
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.,Menzies Institute for Medical Research, Hobart, TAS, Australia
| | - Lingling Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
58
|
WEDAGEDERA JANAKR, BURROUGHS NJ. COMPARISON OF A DUAL STRATEGY FOR T-CELL ACTIVATION UNDER INHIBITION OF THE CD4 RECEPTOR. J BIOL SYST 2018. [DOI: 10.1142/s0218339018500158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We consider a stochastic model for T-cell activation proposed in Refs. [1] and [2] to compare the specificity and sensitivity of two different strategies for T-cell activation that utilize the history of phosphorylation of T-cell receptor (TCR). We compare these two strategies when the temporal signals/events that are essential for progressive T-cell activation are suppressed by blockade of CD4 receptor that may have caused by disease or therapeutic effects.3–6 We show that under these conditions, a threshold-strategy which is capable of maintaining a threshold (for total number of phosphorylated TCRs by time [Formula: see text]) for a further duration [Formula: see text] performs better in discriminating agonist peptides than a single-threshold strategy (reached by time [Formula: see text]) leading to T-cell activation using the Wentzell-Friedlin theory for large deviations for stochastic processes.7,8
Collapse
Affiliation(s)
- JANAK R. WEDAGEDERA
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - N. J. BURROUGHS
- Mathematics Institute and Warwick Systems, Biology Centre, University of Warwick, Coventry CV4 7AL, United Kingdom
| |
Collapse
|
59
|
Xiong W, Chen Y, Kang X, Chen Z, Zheng P, Hsu YH, Jang JH, Qin L, Liu H, Dotti G, Liu D. Immunological Synapse Predicts Effectiveness of Chimeric Antigen Receptor Cells. Mol Ther 2018; 26:963-975. [PMID: 29503199 PMCID: PMC6080133 DOI: 10.1016/j.ymthe.2018.01.020] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 01/19/2018] [Accepted: 01/25/2018] [Indexed: 12/20/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T cell therapy has the potential to improve the overall survival of patients with malignancies by enhancing the effectiveness of CAR T cells. Precisely predicting the effectiveness of various CAR T cells represents one of today’s key unsolved problems in immunotherapy. Here, we predict the effectiveness of CAR-modified cells by evaluating the quality of the CAR-mediated immunological synapse (IS) by quantitation of F-actin, clustering of tumor antigen, polarization of lytic granules (LGs), and distribution of key signaling molecules within the IS. Long-term killing capability, but not secretion of conventional cytokines or standard 4-hr cytotoxicity, correlates positively with the quality of the IS in two different CAR T cells that share identical antigen specificity. Xenograft model data confirm that the quality of the IS in vitro correlates positively with performance of CAR-modified immune cells in vivo. Therefore, we propose that the quality of the IS predicts the effectiveness of CAR-modified immune cells, which provides a novel strategy to guide CAR therapy.
Collapse
MESH Headings
- Animals
- Antigens, CD19/immunology
- Antigens, Neoplasm/immunology
- Biomarkers
- Cell Line
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Gene Expression
- Gene Order
- Genes, Reporter
- Genetic Vectors/genetics
- Humans
- Immunological Synapses/immunology
- Immunological Synapses/metabolism
- Immunotherapy, Adoptive/methods
- Mice
- Microscopy, Confocal
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Retroviridae/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transduction, Genetic
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Wei Xiong
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Yuhui Chen
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Xi Kang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Zhiying Chen
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; Xiangya Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Peilin Zheng
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Yi-Hsin Hsu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Joon Hee Jang
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Hao Liu
- Biostatistics Core of the Dan L. Duncan Cancer Center, Houston, TX 77030, USA
| | - Gianpietro Dotti
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Dongfang Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
60
|
Microfluidic single-cell technology in immunology and antibody screening. Mol Aspects Med 2018; 59:47-61. [DOI: 10.1016/j.mam.2017.09.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 11/20/2022]
|
61
|
Liang W, Mao S, Sun S, Li M, Li Z, Yu R, Ma T, Gu J, Zhang J, Taniguchi N, Li W. Core Fucosylation of the T Cell Receptor Is Required for T Cell Activation. Front Immunol 2018; 9:78. [PMID: 29434598 PMCID: PMC5796888 DOI: 10.3389/fimmu.2018.00078] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/11/2018] [Indexed: 12/05/2022] Open
Abstract
CD4+ T cell activation promotes the pathogenic process of systemic lupus erythematosus (SLE). T cell receptor (TCR) complex are highly core fucosylated glycoproteins, which play important roles in T cell activation. In this study, we found that the core fucosylation of CD4+ T cells was significantly increased in SLE patients. Loss of core fucosyltransferase (Fut8), the sole enzyme for catalyzing the core fucosylation of N-glycan, significantly reduced CD4+ T cell activation and ameliorated the experimental autoimmune encephalomyelitis-induced syndrome in Fut8−/− mice. T cell activation with OVA323–339 loaded major histocompatibility complex II (pMHC-II) on B cell was dramatically attenuated in Fut8−/−OT-II CD4+ T cells compared with Fut8+/+OT-II CD4+ T cells. Moreover, the phosphorylation of ZAP-70 was significantly reduced in Fut8+/+OT-II CD4+ T cells by the treatment of fucosidase. Our results suggest that core fucosylation is required for efficient TCR–pMHC-II contacts in CD4+ T cell activation, and hyper core fucosylation may serve as a potential novel biomarker in the sera from SLE patients.
Collapse
Affiliation(s)
- Wei Liang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Shanshan Mao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Shijie Sun
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ming Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhi Li
- Clinical Laboratory, Dalian Municipal Central Hospital, Dalian, China
| | - Rui Yu
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Jianing Zhang
- School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| | - Naoyuki Taniguchi
- Systems Glycobiology Research Group, Advanced Science Institute, RIKEN, Saitama, Japan
| | - Wenzhe Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
62
|
Hickey JW, Vicente FP, Howard GP, Mao HQ, Schneck JP. Biologically Inspired Design of Nanoparticle Artificial Antigen-Presenting Cells for Immunomodulation. NANO LETTERS 2017; 17:7045-7054. [PMID: 28994285 PMCID: PMC6709596 DOI: 10.1021/acs.nanolett.7b03734] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Particles engineered to engage and interact with cell surface ligands and to modulate cells can be harnessed to explore basic biological questions as well as to devise cellular therapies. Biology has inspired the design of these particles, such as artificial antigen-presenting cells (aAPCs) for use in immunotherapy. While much has been learned about mimicking antigen presenting cell biology, as we decrease the size of aAPCs to the nanometer scale, we need to extend biomimetic design to include considerations of T cell biology-including T-cell receptor (TCR) organization. Here we describe the first quantitative analysis of particle size effect on aAPCs with both Signals 1 and 2 based on T cell biology. We show that aAPCs, larger than 300 nm, activate T cells more efficiently than smaller aAPCs, 50 nm. The 50 nm aAPCs require saturating doses or require artificial magnetic clustering to activate T cells. Increasing ligand density alone on the 50 nm aAPCs did not increase their ability to stimulate CD8+ T cells, confirming the size-dependent phenomenon. These data support the need for multireceptor ligation and activation of T-cell receptor (TCR) nanoclusters of similar sizes to 300 nm aAPCs. Quantitative analysis and modeling of a nanoparticle system provides insight into engineering constraints of aAPCs for T cell immunotherapy applications and offers a case study for other cell-modulating particles.
Collapse
Affiliation(s)
- John W. Hickey
- Department of Biomedical Engineering, School of Medicine
- Institute for Cell Engineering, School of Medicine
- Translational Tissue Engineering Center
- Institute for Nanobiotechnology
| | | | - Gregory P. Howard
- Department of Biomedical Engineering, School of Medicine
- Institute for Nanobiotechnology
| | - Hai-Quan Mao
- Translational Tissue Engineering Center
- Institute for Nanobiotechnology
- Department of Materials Science and Engineering, Whiting School of Engineering
| | - Jonathan P. Schneck
- Institute for Cell Engineering, School of Medicine
- Department of Pathology, School of Medicine
- Institute for Nanobiotechnology
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Corresponding Author: . Phone: 410-614-4589
| |
Collapse
|
63
|
Baer A, Colon-Moran W, Xiang J, Stapleton JT, Bhattarai N. Src-family kinases negatively regulate NFAT signaling in resting human T cells. PLoS One 2017; 12:e0187123. [PMID: 29073235 PMCID: PMC5658144 DOI: 10.1371/journal.pone.0187123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/13/2017] [Indexed: 01/30/2023] Open
Abstract
T cell signaling is required for activation of both natural and therapeutic T cells including chimeric antigen receptor (CAR) T cells. Identification of novel factors and pathways regulating T cell signaling may aid in development of effective T cell therapies. In resting human T cells, the majority of Src-family of tyrosine kinases (SFKs) are inactive due to phosphorylation of a conserved carboxy-terminal tyrosine residue. Recently, a pool of enzymatically active SFKs has been identified in resting T cells; however, the significance of these is incompletely understood. Here, we characterized the role of active SFKs in resting human T cells. Pharmacologic inhibition of active SFKs enhanced distal TCR signaling as measured by IL-2 release and CD25 surface expression following TCR-independent activation. Mechanistically, inhibition of the active pool of SFKs induced nuclear translocation of NFAT1, and enhanced NFAT1-dependent signaling in resting T cells. The negative regulation of NFAT1 signaling was in part mediated by the Src-kinase Lck as human T cells lacking Lck had increased levels of nuclear NFAT1 and demonstrated enhanced NFAT1-dependent gene expression. Inhibition of active SFKs in resting primary human T cells also increased nuclear NFAT1 and enhanced NFAT1-dependent signaling. Finally, the calcineurin inhibitor FK506 and Cyclosporin A reversed the effect of SFKs inhibition on NFAT1. Together, these data identified a novel role of SFKs in preventing aberrant NFAT1 activation in resting T cells, and suggest that maintaining this pool of active SFKs in therapeutic T cells may increase the efficacy of T cell therapies.
Collapse
Affiliation(s)
- Alan Baer
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Winston Colon-Moran
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Jinhua Xiang
- Research Service, Iowa City Veterans Affairs Medical Center, and the Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Jack T. Stapleton
- Research Service, Iowa City Veterans Affairs Medical Center, and the Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Nirjal Bhattarai
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
- * E-mail:
| |
Collapse
|
64
|
Udenwobele DI, Su RC, Good SV, Ball TB, Varma Shrivastav S, Shrivastav A. Myristoylation: An Important Protein Modification in the Immune Response. Front Immunol 2017; 8:751. [PMID: 28713376 PMCID: PMC5492501 DOI: 10.3389/fimmu.2017.00751] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/13/2017] [Indexed: 01/24/2023] Open
Abstract
Protein N-myristoylation is a cotranslational lipidic modification specific to the alpha-amino group of an N-terminal glycine residue of many eukaryotic and viral proteins. The ubiquitous eukaryotic enzyme, N-myristoyltransferase, catalyzes the myristoylation process. Precisely, attachment of a myristoyl group increases specific protein–protein interactions leading to subcellular localization of myristoylated proteins with its signaling partners. The birth of the field of myristoylation, a little over three decades ago, has led to the understanding of the significance of protein myristoylation in regulating cellular signaling pathways in several biological processes especially in carcinogenesis and more recently immune function. This review discusses myristoylation as a prerequisite step in initiating many immune cell signaling cascades. In particular, we discuss the hitherto unappreciated implication of myristoylation during myelopoiesis, innate immune response, lymphopoiesis for T cells, and the formation of the immunological synapse. Furthermore, we discuss the role of myristoylation in inducing the virological synapse during human immunodeficiency virus infection as well as its clinical implication. This review aims to summarize existing knowledge in the field and to highlight gaps in our understanding of the role of myristoylation in immune function so as to further investigate into the dynamics of myristoylation-dependent immune regulation.
Collapse
Affiliation(s)
- Daniel Ikenna Udenwobele
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada.,Department of Biochemistry, University of Nigeria, Nsukka, Enugu, Nigeria
| | - Ruey-Chyi Su
- JC Wilt Infectious Diseases Research Institute, National HIV and Retrovirology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Sara V Good
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
| | - Terry Blake Ball
- JC Wilt Infectious Diseases Research Institute, National HIV and Retrovirology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Shailly Varma Shrivastav
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada.,VastCon Inc., Winnipeg, MB, Canada
| | - Anuraag Shrivastav
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
65
|
Nanoparticles for tumor immunotherapy. Eur J Pharm Biopharm 2017; 115:243-256. [DOI: 10.1016/j.ejpb.2017.03.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 03/01/2017] [Accepted: 03/17/2017] [Indexed: 12/21/2022]
|
66
|
Liu D, Tian S, Zhang K, Xiong W, Lubaki NM, Chen Z, Han W. Chimeric antigen receptor (CAR)-modified natural killer cell-based immunotherapy and immunological synapse formation in cancer and HIV. Protein Cell 2017; 8:861-877. [PMID: 28488245 PMCID: PMC5712291 DOI: 10.1007/s13238-017-0415-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/22/2017] [Indexed: 12/31/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells contribute to the body’s immune defenses. Current chimeric antigen receptor (CAR)-modified T cell immunotherapy shows strong promise for treating various cancers and infectious diseases. Although CAR-modified NK cell immunotherapy is rapidly gaining attention, its clinical applications are mainly focused on preclinical investigations using the NK92 cell line. Despite recent advances in CAR-modified T cell immunotherapy, cost and severe toxicity have hindered its widespread use. To alleviate these disadvantages of CAR-modified T cell immunotherapy, additional cytotoxic cell-mediated immunotherapies are urgently needed. The unique biology of NK cells allows them to serve as a safe, effective, alternative immunotherapeutic strategy to CAR-modified T cells in the clinic. While the fundamental mechanisms underlying the cytotoxicity and side effects of CAR-modified T and NK cell immunotherapies remain poorly understood, the formation of the immunological synapse (IS) between CAR-modified T or NK cells and their susceptible target cells is known to be essential. The role of the IS in CAR T and NK cell immunotherapies will allow scientists to harness the power of CAR-modified T and NK cells to treat cancer and infectious diseases. In this review, we highlight the potential applications of CAR-modified NK cells to treat cancer and human immunodeficiency virus (HIV), and discuss the challenges and possible future directions of CAR-modified NK cell immunotherapy, as well as the importance of understanding the molecular mechanisms of CAR-modified T cell- or NK cell-mediated cytotoxicity and side effects, with a focus on the CAR-modified NK cell IS.
Collapse
Affiliation(s)
- Dongfang Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA.
| | - Shuo Tian
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kai Zhang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Wei Xiong
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Ndongala Michel Lubaki
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Zhiying Chen
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Weidong Han
- Institute of Basic Medicine, College of Life Sciences, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
67
|
In vitro tracking and intracellular protein distribution in immunology. Immunol Cell Biol 2017; 95:501-505. [PMID: 28392557 DOI: 10.1038/icb.2017.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 03/27/2017] [Accepted: 04/04/2017] [Indexed: 11/08/2022]
Abstract
New imaging techniques have enabled major advances in understanding how immune reactions are initiated, coordinated and controlled. Imaging methods, which were previously mostly descriptive and supplementary to more quantitative approaches, have now reached sufficient precision and throughput that they are becoming integral to almost all aspects of immunology research. Imaging methodologies that increase the resolution and sensitivity of detection, alongside an ever-expanding range of fluorescent reporters of molecular and cellular activity, and vastly improved analysis methods, have all facilitated this transformation. In this review, we will discuss how advances in imaging are changing the way we view immune activation and control using T cells as the model immune system. We will describe how imaging has transformed our knowledge of molecular and signalling events in T-cell activation, and the impact of these molecular events on the behaviour of T cells.
Collapse
|
68
|
Yi Y, Sanchez L, Gao Y, Lee K, Yu Y. Interrogating Cellular Functions with Designer Janus Particles. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2017; 29:1448-1460. [PMID: 31530969 PMCID: PMC6748339 DOI: 10.1021/acs.chemmater.6b05322] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Janus particles have two distinct surfaces or compartments. This enables novel applications that are impossible with homogeneous particles, ranging from the engineering of active colloidal metastructures to creating multimodal therapeutic materials. Recent years have witnessed a rapid development of novel Janus structures and exploration of their applications, particularly in the biomedical arena. It, therefore, becomes crucial to understand how Janus particles with surface or structural anisotropy might interact with biological systems and how such interactions may be exploited to manipulate biological responses. This perspective highlights recent studies that have employed Janus particles as novel toolsets to manipulate, measure, and understand cellular functions. Janus particles have been shown to have biological interactions different from uniform particles. Their surface anisotropy has been used to control the cell entry of synthetic particles, to spatially organize stimuli for the activation of immune cells, and to enable direct visualization and measurement of rotational dynamics of particles in living systems. The work included in this perspective showcases the significance of understanding the biological interactions of Janus particles and the tremendous potential of harnessing such interactions to advance the development of Janus structure-based biomaterials.
Collapse
Affiliation(s)
| | | | | | | | - Yan Yu
- Corresponding Author (Y.Yu)
| |
Collapse
|
69
|
Kramer K, Young SL, Walker GF. Comparative Study of 5'- and 3'-Linked CpG-Antigen Conjugates for the Induction of Cellular Immune Responses. ACS OMEGA 2017; 2:227-235. [PMID: 30023513 PMCID: PMC6044624 DOI: 10.1021/acsomega.6b00368] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 01/12/2017] [Indexed: 06/08/2023]
Abstract
Conjugation of CpG to an antigen induces a stronger immune response compared to that of the mixture. This study compares the in vitro immunostimulatory activity of CpG conjugated via either its 5' or 3' end to the model antigen ovalbumin (OVA). CpG modified with an amine at either the 5' or 3' end was conjugated to OVA via a stable bis-aryl hydrazone bond. Similar levels of CpG conjugation to OVA were observed for both conjugates on the basis of the absorbance at 360 nm for the formation of the bis-aryl hydrazone bond, which determined 2.8 ± 0.3 CpGs linked per OVA. Both the 5' and 3' CpG-OVA conjugates had similar size-exclusion chromatography elution profiles. The immunostimulatory properties of the conjugates were determined by dendritic cells (DCs) and T-cells isolated from mice. The activation of DCs was determined by the upregulation of activation markers CD86 and CD40. T-cells were co-cultured with stimulated DCs, and the immunogenicity was determined by measuring T-cell proliferation and interferon γ production. Both the CpG 5'- and 3'-linked conjugates induced the same level (p > 0.5) of DC activation markers, which were significantly higher than those of the untreated control. Similarly, T-cell assays showed no significant difference (p > 0.5) between the 5' and 3' conjugates with respect to T-cell proliferation and interferon γ production. The 5' and 3' conjugates induced T-cell activation significantly higher than the mixture of CpG and OVA. This study showed that the end at which CpG is conjugated to an antigen has no influence on the generation of a T-cell-based immune response in vitro.
Collapse
Affiliation(s)
- Katrin Kramer
- School
of Pharmacy and Department of Pathology, Dunedin School of Medicine, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand
| | - Sarah L. Young
- School
of Pharmacy and Department of Pathology, Dunedin School of Medicine, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand
| | - Greg F. Walker
- School
of Pharmacy and Department of Pathology, Dunedin School of Medicine, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand
| |
Collapse
|
70
|
Barr VA, Sherman E, Yi J, Akpan I, Rouquette-Jazdanian AK, Samelson LE. Development of nanoscale structure in LAT-based signaling complexes. J Cell Sci 2016; 129:4548-4562. [PMID: 27875277 PMCID: PMC5201021 DOI: 10.1242/jcs.194886] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/04/2016] [Indexed: 12/30/2022] Open
Abstract
The adapter molecule linker for activation of T cells (LAT) plays a crucial role in forming signaling complexes induced by stimulation of the T cell receptor (TCR). These multi-molecular complexes are dynamic structures that activate highly regulated signaling pathways. Previously, we have demonstrated nanoscale structure in LAT-based complexes where the adapter SLP-76 (also known as LCP2) localizes to the periphery of LAT clusters. In this study, we show that initially LAT and SLP-76 are randomly dispersed throughout the clusters that form upon TCR engagement. The segregation of LAT and SLP-76 develops near the end of the spreading process. The local concentration of LAT also increases at the same time. Both changes require TCR activation and an intact actin cytoskeleton. These results demonstrate that the nanoscale organization of LAT-based signaling complexes is dynamic and indicates that different kinds of LAT-based complexes appear at different times during T cell activation.
Collapse
Affiliation(s)
- Valarie A Barr
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| | - Jason Yi
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Itoro Akpan
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | | | - Lawrence E Samelson
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
71
|
Blas-Rus N, Bustos-Morán E, Martín-Cófreces NB, Sánchez-Madrid F. Aurora-A shines on T cell activation through the regulation of Lck. Bioessays 2016; 39. [PMID: 27910998 DOI: 10.1002/bies.201600156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Different protein kinases control signaling emanating from the T cell receptor (TCR) during antigen-specific T cell activation. Mitotic kinases, e.g. Aurora-A, have been widely studied in the context of mitosis due to their role during microtubule (MT) nucleation, becoming critical regulators of cell cycle progression. We have recently described a specific role for Aurora-A kinase in antigenic T cell activation. Blockade of Aurora-A in T cells severely disrupts the dynamics of MTs and CD3ζ-bearing signaling vesicles during T cell activation. Furthermore, Aurora-A deletion impairs the activation of signaling molecules downstream of the TCR. Targeting Aurora-A disturbs the activation of Lck, which is one of the first signals that drive T cell activation in an antigen-dependent manner. This work describes possible models of regulation of Lck by Aurora-A during T cell activation. We also discuss possible roles for Aurora-A in other systems similar to the IS, and its putative functions in cell polarization.
Collapse
Affiliation(s)
- Noelia Blas-Rus
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain
| | - Eugenio Bustos-Morán
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Noa B Martín-Cófreces
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain.,Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
72
|
Ballek O, Valečka J, Dobešová M, Broučková A, Manning J, Řehulka P, Stulík J, Filipp D. TCR Triggering Induces the Formation of Lck-RACK1-Actinin-1 Multiprotein Network Affecting Lck Redistribution. Front Immunol 2016; 7:449. [PMID: 27833610 PMCID: PMC5081367 DOI: 10.3389/fimmu.2016.00449] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/10/2016] [Indexed: 02/02/2023] Open
Abstract
The initiation of T-cell signaling is critically dependent on the function of the member of Src family tyrosine kinases, Lck. Upon T-cell antigen receptor (TCR) triggering, Lck kinase activity induces the nucleation of signal-transducing hubs that regulate the formation of complex signaling network and cytoskeletal rearrangement. In addition, the delivery of Lck function requires rapid and targeted membrane redistribution, but the mechanism underpinning this process is largely unknown. To gain insight into this process, we considered previously described proteins that could assist in this process via their capacity to interact with kinases and regulate their intracellular translocations. An adaptor protein, receptor for activated C kinase 1 (RACK1), was chosen as a viable option, and its capacity to bind Lck and aid the process of activation-induced redistribution of Lck was assessed. Our microscopic observation showed that T-cell activation induces a rapid, concomitant, and transient co-redistribution of Lck and RACK1 into the forming immunological synapse. Consistent with this observation, the formation of transient RACK1-Lck complexes were detectable in primary CD4+ T-cells with their maximum levels peaking 10 s after TCR-CD4 co-aggregation. Moreover, RACK1 preferentially binds to a pool of kinase active pY394Lck, which co-purifies with high molecular weight cellular fractions. The formation of RACK1-Lck complexes depends on functional SH2 and SH3 domains of Lck and includes several other signaling and cytoskeletal elements that transiently bind the complex. Notably, the F-actin-crosslinking protein, α-actinin-1, binds to RACK1 only in the presence of kinase active Lck suggesting that the formation of RACK1-pY394Lck-α-actinin-1 complex serves as a signal module coupling actin cytoskeleton bundling with productive TCR/CD4 triggering. In addition, the treatment of CD4+ T-cells with nocodazole, which disrupts the microtubular network, also blocked the formation of RACK1-Lck complexes. Importantly, activation-induced Lck redistribution was diminished in primary CD4+ T-cells by an adenoviral-mediated knockdown of RACK1. These results demonstrate that in T cells, RACK1, as an essential component of the multiprotein complex which upon TCR engagement, links the binding of kinase active Lck to elements of the cytoskeletal network and affects the subcellular redistribution of Lck.
Collapse
Affiliation(s)
- Ondřej Ballek
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Jan Valečka
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Martina Dobešová
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Adéla Broučková
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Jasper Manning
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Pavel Řehulka
- Faculty of Military Health Sciences, Institute of Molecular Pathology , Hradec Králové , Czech Republic
| | - Jiří Stulík
- Faculty of Military Health Sciences, Institute of Molecular Pathology , Hradec Králové , Czech Republic
| | - Dominik Filipp
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| |
Collapse
|
73
|
Lee HW, Gangadaran P, Kalimuthu S, Ahn BC. Advances in Molecular Imaging Strategies for In Vivo Tracking of Immune Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1946585. [PMID: 27725934 PMCID: PMC5048043 DOI: 10.1155/2016/1946585] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/12/2016] [Accepted: 08/23/2016] [Indexed: 01/25/2023]
Abstract
Tracking of immune cells in vivo is a crucial tool for development and optimization of cell-based therapy. Techniques for tracking immune cells have been applied widely for understanding the intrinsic behavior of immune cells and include non-radiation-based techniques such as optical imaging and magnetic resonance imaging (MRI), radiation-based techniques such as computerized tomography (CT), and nuclear imaging including single photon emission computerized tomography (SPECT) and positron emission tomography (PET). Each modality has its own strengths and limitations. To overcome the limitations of each modality, multimodal imaging techniques involving two or more imaging modalities are actively applied. Multimodal techniques allow integration of the strengths of individual modalities. In this review, we discuss the strengths and limitations of currently available preclinical in vivo immune cell tracking techniques and summarize the value of immune cell tracking in the development and optimization of immune cell therapy for various diseases.
Collapse
Affiliation(s)
- Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| |
Collapse
|
74
|
A major secretory defect of tumour-infiltrating T lymphocytes due to galectin impairing LFA-1-mediated synapse completion. Nat Commun 2016; 7:12242. [PMID: 27447355 PMCID: PMC4961845 DOI: 10.1038/ncomms12242] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 06/15/2016] [Indexed: 12/20/2022] Open
Abstract
Surface galectin has been shown to contribute to dysfunctions of human tumour-infiltrating lymphocytes (TILs). We show here that galectin-covered CD8 TILs produce normal amounts of intracellular cytokines, but fail to secrete them because of defective actin rearrangements at the synapse. The non-secreting TILs also display reduced adhesion to their targets, together with defective LFA-1 recruitment and activation at the synapse. These defects are relieved by releasing surface galectin. As mild LFA-1 blockade on normal blood T cells emulate the defects of galectin-covered TILs, we conclude that galectin prevents the formation of a functional secretory synapse by preventing optimal LFA-1 triggering. Our results highlight a major secretory defect of TILs that is not revealed by widely used intracellular cytokine immunomonitoring assays. They also provide additional insights into the T-cell response, by showing that different thresholds of LFA-1 triggering are required to promote the intracellular production of cytokines and their secretion. Galectin-3 is a sugar-binding protein that can inhibit antitumour cytotoxic immunity. Here the authors show that Galectin-3 expressed by tumour cells inhibits LFA-1 on cytotoxic lymphocytes, impairing immunological synapse formation, IFNg secretion, and target cell killing.
Collapse
|
75
|
Hashimoto-Tane A, Saito T. Dynamic Regulation of TCR-Microclusters and the Microsynapse for T Cell Activation. Front Immunol 2016; 7:255. [PMID: 27446085 PMCID: PMC4923147 DOI: 10.3389/fimmu.2016.00255] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 06/15/2016] [Indexed: 11/13/2022] Open
Abstract
The interaction between a T cell and an antigen-presenting cell is the initiating event in T cell-mediated adaptive immunity. The Immunological Synapse (IS) is formed at the interface between these two cell types, and is the site where antigen (Ag)-specific recognition and activation are induced through the T cell receptor (TCR). This occurs at the center of the IS, and cell adhesion is supported through integrins in the area surrounding the TCR. Recently, this model has been revised based on data indicating that the initial Ag-specific activation signal is triggered prior to IS formation at TCR-microclusters (MCs), sites where TCR, kinases and adaptors of TCR proximal downstream signaling molecules accumulate as an activation signaling cluster. TCR-MCs then move into the center of the cell-cell interface to generate the cSMAC. This translocation of TCR-MCs is mediated initially by the actin cytoskeleton and then by dynein-induced movement along microtubules. The translocation of TCR-MCs and cSMAC formation is induced upon strong TCR stimulation through the assembly of a TCR-dynein super complex with microtubules. The Ag-specific activation signal is induced at TCR-MCs, but the adhesion signal is now shown to be induced by generating a "microsynapse," which is composed of a core of TCR-MCs and the surrounding adhesion ring of integrin and focal adhesion molecules. Since the microsynapse is critical for activation, particularly under weak TCR stimulation, this structure supports a weak TCR signal through a cell-cell adhesion signal. The microsynapse has a structure similar to the IS but on a micro-scale and regulates Ag-specific activation as well as cell-cell adhesion. We describe here the dynamic regulation of TCR-MCs, responsible for inducing Ag-specific activation signals, and the microsynapse, responsible for adhesion signals critical for cell-cell interactions, and their interrelationship.
Collapse
Affiliation(s)
- Akiko Hashimoto-Tane
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences , Yokohama , Japan
| | - Takashi Saito
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences , Yokohama , Japan
| |
Collapse
|
76
|
DNA-based nanoparticle tension sensors reveal that T-cell receptors transmit defined pN forces to their antigens for enhanced fidelity. Proc Natl Acad Sci U S A 2016; 113:5610-5. [PMID: 27140637 DOI: 10.1073/pnas.1600163113] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
T cells are triggered when the T-cell receptor (TCR) encounters its antigenic ligand, the peptide-major histocompatibility complex (pMHC), on the surface of antigen presenting cells (APCs). Because T cells are highly migratory and antigen recognition occurs at an intermembrane junction where the T cell physically contacts the APC, there are long-standing questions of whether T cells transmit defined forces to their TCR complex and whether chemomechanical coupling influences immune function. Here we develop DNA-based gold nanoparticle tension sensors to provide, to our knowledge, the first pN tension maps of individual TCR-pMHC complexes during T-cell activation. We show that naïve T cells harness cytoskeletal coupling to transmit 12-19 pN of force to their TCRs within seconds of ligand binding and preceding initial calcium signaling. CD8 coreceptor binding and lymphocyte-specific kinase signaling are required for antigen-mediated cell spreading and force generation. Lymphocyte function-associated antigen 1 (LFA-1) mediated adhesion modulates TCR-pMHC tension by intensifying its magnitude to values >19 pN and spatially reorganizes the location of TCR forces to the kinapse, the zone located at the trailing edge of migrating T cells, thus demonstrating chemomechanical crosstalk between TCR and LFA-1 receptor signaling. Finally, T cells display a dampened and poorly specific response to antigen agonists when TCR forces are chemically abolished or physically "filtered" to a level below ∼12 pN using mechanically labile DNA tethers. Therefore, we conclude that T cells tune TCR mechanics with pN resolution to create a checkpoint of agonist quality necessary for specific immune response.
Collapse
|
77
|
Abstract
Human immunodeficiency virus type 1 (HIV-1) gives rise to a chronic infection that progressively depletes CD4(+) T lymphocytes. CD4(+) T lymphocytes play a central coordinating role in adaptive cellular and humoral immune responses, and to do so they migrate and interact within lymphoid compartments and at effector sites to mount immune responses. While cell-free virus serves as an excellent prognostic indicator for patient survival, interactions of infected T cells or virus-scavenging immune cells with uninfected T cells can greatly enhance viral spread. HIV can induce interactions between infected and uninfected T cells that are triggered by cell surface expression of viral Env, which serves as a cell adhesion molecule that interacts with CD4 on the target cell, before it acts as the viral membrane fusion protein. These interactions are called virological synapses and promote replication in the face of selective pressure of humoral immune responses and antiretroviral therapy. Other infection-enhancing cell-cell interactions occur between virus-concentrating antigen-presenting cells and recipient T cells, called infectious synapses. The exact roles that these cell-cell interactions play in each stage of infection, from viral acquisition, systemic dissemination, to chronic persistence are still being determined. Infection-promoting immune cell interactions are likely to contribute to viral persistence and enhance the ability of HIV-1 to evade adaptive immune responses.
Collapse
Affiliation(s)
- K M Law
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - N Satija
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - A M Esposito
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - B K Chen
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
78
|
Cush SS, Reynoso GV, Kamenyeva O, Bennink JR, Yewdell JW, Hickman HD. Locally Produced IL-10 Limits Cutaneous Vaccinia Virus Spread. PLoS Pathog 2016; 12:e1005493. [PMID: 26991092 PMCID: PMC4798720 DOI: 10.1371/journal.ppat.1005493] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 02/15/2016] [Indexed: 12/29/2022] Open
Abstract
Skin infection with the poxvirus vaccinia (VV) elicits a powerful, inflammatory cellular response that clears virus infection in a coordinated, spatially organized manner. Given the high concentration of pro-inflammatory effectors at areas of viral infection, it is unclear how tissue pathology is limited while virus-infected cells are being eliminated. To better understand the spatial dynamics of the anti-inflammatory response to a cutaneous viral infection, we first screened cytokine mRNA expression levels after epicutaneous (ec.) VV infection and found a large increase the anti-inflammatory cytokine IL-10. Ex vivo analyses revealed that T cells in the skin were the primary IL-10-producing cells. To understand the distribution of IL-10-producing T cells in vivo, we performed multiphoton intravital microscopy (MPM) of VV-infected mice, assessing the location and dynamic behavior of IL-10 producing cells. Although virus-specific T cells were distributed throughout areas of the inflamed skin lacking overt virus-infection, IL-10+ cells closely associated with large keratinocytic foci of virus replication where they exhibited similar motility patterns to bulk antigen-specific CD8+ T cells. Paradoxically, neutralizing secreted IL-10 in vivo with an anti-IL-10 antibody increased viral lesion size and viral replication. Additional analyses demonstrated that IL-10 antibody administration decreased recruitment of CCR2+ inflammatory monocytes, which were important for reducing viral burden in the infected skin. Based upon these findings, we conclude that spatially concentrated IL-10 production limits cutaneous viral replication and dissemination, likely through modulation of the innate immune repertoire at the site of viral growth.
Collapse
Affiliation(s)
- Stephanie S. Cush
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Glennys V. Reynoso
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jack R. Bennink
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jonathan W. Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Heather D. Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
79
|
Carlson A, Mahadevan L. Elastohydrodynamics and Kinetics of Protein Patterning in the Immunological Synapse. PLoS Comput Biol 2015; 11:e1004481. [PMID: 26699430 PMCID: PMC4689476 DOI: 10.1371/journal.pcbi.1004481] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 07/30/2015] [Indexed: 11/19/2022] Open
Abstract
We propose a minimal mathematical model for the physical basis of membrane protein patterning in the immunological synapse (IS), which encompass membrane mechanics, protein binding kinetics and motion, and fluid flow in the synaptic cleft. Our theory leads to simple predictions for the spatial and temporal scales of protein cluster formation, growth and arrest as a function of membrane stiffness, rigidity and kinetics of the adhesive proteins, and the fluid flow in the synaptic cleft. Numerical simulations complement these scaling laws by quantifying the nucleation, growth and stabilization of proteins domains on the size of the cell. Direct comparison with experiment shows that passive elastohydrodynamics and kinetics of protein binding in the synaptic cleft can describe the short-time formation and organization of protein clusters, without evoking any active processes in the cytoskeleton. Despite the apparent complexity of the process, our analysis shows that just two dimensionless parameters characterize the spatial and temporal evolution of the protein pattern: a ratio of membrane elasticity to protein stiffness, and the ratio of a hydrodynamic time scale for fluid flow relative to the protein binding rate. A simple phase diagram encompasses the variety of patterns that can arise.
Collapse
Affiliation(s)
- Andreas Carlson
- School of Engineering and Applied Sciences, Kavli Institute for Bionano Science and Technology, and Wyss Institute, Harvard University, Cambridge, United States of America
| | - L. Mahadevan
- School of Engineering and Applied Sciences, Kavli Institute for Bionano Science and Technology, and Wyss Institute, Harvard University, Cambridge, United States of America
- Departments of Physics, and Organismic and Evolutionary Biology, Harvard University, Cambridge, United States of America
- * E-mail:
| |
Collapse
|
80
|
Impact of Leukocyte Function-Associated Antigen-1 Blockade on Endogenous Allospecific T Cells to Multiple Minor Histocompatibility Antigen Mismatched Cardiac Allograft. Transplantation 2015; 99:2485-93. [DOI: 10.1097/tp.0000000000000805] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
81
|
Badawi AH, Kiptoo P, Siahaan TJ. Immune Tolerance Induction against Experimental Autoimmune Encephalomyelitis (EAE) Using A New PLP-B7AP Conjugate that Simultaneously Targets B7/CD28 Costimulatory Signal and TCR/MHC-II Signal. JOURNAL OF MULTIPLE SCLEROSIS 2015; 2:1000131. [PMID: 26140285 PMCID: PMC4484621 DOI: 10.4172/2376-0389.1000131] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Most of the current therapies used in the treatment of multiple sclerosis (MS) are either ineffective or have adverse side effects. As such, there is a need to develop better therapies that specifically target myelin-specific aberrant immune cells involved in CNS inflammation without compromising the general immune system. In the present study, we developed a new bifunctional peptide inhibitor (BPI) that is effective and specific. Our BPI (PLP-B7AP) is composed of an antigenic peptide from myelin proteolipid protein (PLP139-151) and a B7 antisense peptide (B7AP) derived from CD28 receptor. The main hypothesis is that PLP-B7AP simultaneously targets MHC-II and B7-costimulatory molecules on the surface of antigen presenting cells (APC) and possibly alters the differentiation of naïve T cells from inflammatory to regulatory phenotypes. Results showed that PLP-B7AP was very effective in suppressing experimental autoimmune encephalomyelitis (EAE) compared to various controls in a mouse model. PLP-B7AP was effective when administered both before and after disease induction. Secreted cytokines from splenocytes isolated during periods of high disease severity and remission indicated that PLP-B7AP treatment induced an increased production of anti-inflammatory cytokines and inhibited the production of pro-inflammatory cytokines. Further, analysis of cortical brain tissue sections showed that PLP-B7AP treated mice had significantly lower demyelination compared to the control group. All these taken together indicate that the T cell receptor (TCR) and the CD28 receptor can be targeted simultaneously to improve efficacy and specificity of potential MS therapeutics.
Collapse
Affiliation(s)
- Ahmed H Badawi
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66047, USA
- KU Medical Center, The University of Kansas, Kansas City, KS 66160, USA
| | - Paul Kiptoo
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66047, USA
| | - Teruna J Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
82
|
Wang H, Morita CT. Sensor Function for Butyrophilin 3A1 in Prenyl Pyrophosphate Stimulation of Human Vγ2Vδ2 T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:4583-94. [PMID: 26475929 DOI: 10.4049/jimmunol.1500314] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 09/15/2015] [Indexed: 11/19/2022]
Abstract
Vγ2Vδ2 T cells play important roles in human immunity to pathogens and in cancer immunotherapy by responding to isoprenoid metabolites, such as (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate and isopentenyl pyrophosphate. The Ig superfamily protein butyrophilin (BTN)3A1 was shown to be required for prenyl pyrophosphate stimulation. We proposed that the intracellular B30.2 domain of BTN3A1 binds prenyl pyrophosphates, resulting in a change in the extracellular BTN3A1 dimer that is detected by Vγ2Vδ2 TCRs. Such B30.2 binding was demonstrated recently. However, other investigators reported that the extracellular BTN3A1 IgV domain binds prenyl pyrophosphates, leading to the proposal that the Vγ2Vδ2 TCR recognizes the complex. To distinguish between these mechanisms, we mutagenized residues in the two binding sites and tested the mutant BTN3A1 proteins for their ability to mediate prenyl pyrophosphate stimulation of Vγ2Vδ2 T cells to proliferate and secrete TNF-α. Mutagenesis of residues in the IgV site had no effect on Vγ2Vδ2 T cell proliferation or secretion of TNF-α. In contrast, mutagenesis of residues within the basic pocket and surrounding V regions of the B30.2 domain abrogated prenyl pyrophosphate-induced proliferation. Mutations of residues making hydrogen bonds to the pyrophosphate moiety also abrogated TNF-α secretion, as did mutation of aromatic residues making contact with the alkenyl chain. Some mutations further from the B30.2 binding site also diminished stimulation, suggesting that the B30.2 domain may interact with a second protein. These findings support intracellular sensing of prenyl pyrophosphates by BTN3A1 rather than extracellular presentation.
Collapse
Affiliation(s)
- Hong Wang
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246; and
| | - Craig T Morita
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246; and Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| |
Collapse
|
83
|
Jang JH, Huang Y, Zheng P, Jo MC, Bertolet G, Zhu MX, Qin L, Liu D. Imaging of Cell-Cell Communication in a Vertical Orientation Reveals High-Resolution Structure of Immunological Synapse and Novel PD-1 Dynamics. THE JOURNAL OF IMMUNOLOGY 2015; 195:1320-30. [PMID: 26123352 DOI: 10.4049/jimmunol.1403143] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/23/2015] [Indexed: 12/22/2022]
Abstract
The immunological synapse (IS) is one of the most pivotal communication strategies in immune cells. Understanding the molecular basis of the IS provides critical information regarding how immune cells mount an effective immune response. Fluorescence microscopy provides a fundamental tool to study the IS. However, current imaging techniques for studying the IS cannot sufficiently achieve high resolution in real cell-cell conjugates. In this study, we present a new device that allows for high-resolution imaging of the IS with conventional confocal microscopy in a high-throughput manner. Combining micropits and single-cell trap arrays, we have developed a new microfluidic platform that allows visualization of the IS in vertically "stacked" cells. Using this vertical cell pairing (VCP) system, we investigated the dynamics of the inhibitory synapse mediated by an inhibitory receptor, programed death protein-1, and the cytotoxic synapse at the single-cell level. In addition to the technique innovation, we have demonstrated novel biological findings by this VCP device, including novel distribution of F-actin and cytolytic granules at the IS, programed death protein-1 microclusters at the NK IS, and kinetics of cytotoxicity. We propose that this high-throughput, cost-effective, easy-to-use VCP system, along with conventional imaging techniques, can be used to address a number of significant biological questions in a variety of disciplines.
Collapse
Affiliation(s)
- Joon Hee Jang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030; Center for Human Immunobiology, Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Yu Huang
- Center for Human Immunobiology, Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030; Department of Integrative Biology and Pharmacology, Graduate Program in Cell and Regulatory Biology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Peilin Zheng
- Center for Human Immunobiology, Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Myeong Chan Jo
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030
| | - Grant Bertolet
- Center for Human Immunobiology, Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030; and
| | - Michael Xi Zhu
- Department of Integrative Biology and Pharmacology, Graduate Program in Cell and Regulatory Biology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065
| | - Dongfang Liu
- Center for Human Immunobiology, Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030; and
| |
Collapse
|
84
|
Dinic J, Riehl A, Adler J, Parmryd I. The T cell receptor resides in ordered plasma membrane nanodomains that aggregate upon patching of the receptor. Sci Rep 2015; 5:10082. [PMID: 25955440 PMCID: PMC5386217 DOI: 10.1038/srep10082] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/27/2015] [Indexed: 01/17/2023] Open
Abstract
Two related models for T cell signalling initiation suggest either that T cell receptor (TCR) engagement leads to its recruitment to ordered membrane domains, often referred to as lipid rafts, where signalling molecules are enriched or that ordered TCR-containing membrane nanodomains coalesce upon TCR engagement. That ordered domains form upon TCR engagement, as they do upon lipid raft marker patching, has not been considered. The target of this study was to differentiate between those three options. Plasma membrane order was followed in live T cells at 37 °C using laurdan to report on lipid packing. Patching of the TCR that elicits a signalling response resulted in aggregation, not formation, of ordered plasma membrane domains in both Jurkat and primary T cells. The TCR colocalised with actin filaments at the plasma membrane in unstimulated Jurkat T cells, consistent with it being localised to ordered membrane domains. The colocalisation was most prominent in cells in G1 phase when the cells are ready to commit to proliferation. At other cell cycle phases the TCR was mainly found at perinuclear membranes. Our study suggests that the TCR resides in ordered plasma membrane domains that are linked to actin filaments and aggregate upon TCR engagement.
Collapse
Affiliation(s)
- Jelena Dinic
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| | - Astrid Riehl
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| | - Jeremy Adler
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| | - Ingela Parmryd
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| |
Collapse
|
85
|
Perica K, Kosmides AK, Schneck JP. Linking form to function: Biophysical aspects of artificial antigen presenting cell design. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:781-90. [PMID: 25200637 PMCID: PMC4344884 DOI: 10.1016/j.bbamcr.2014.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/15/2014] [Accepted: 09/01/2014] [Indexed: 12/22/2022]
Abstract
Artificial antigen presenting cells (aAPCs) are engineered platforms for T cell activation and expansion, synthesized by coupling T cell activating proteins to the surface of cell lines or biocompatible particles. They can serve both as model systems to study the basic aspects of T cell signaling and translationally as novel approaches for either active or adoptive immunotherapy. Historically, these reductionist systems have not been designed to mimic the temporally and spatially complex interactions observed during endogenous T cell-APC contact, which include receptor organization at both micro- and nanoscales and dynamic changes in cell and membrane morphologies. Here, we review how particle size and shape, as well as heterogenous distribution of T cell activating proteins on the particle surface, are critical aspects of aAPC design. In doing so, we demonstrate how insights derived from endogenous T cell activation can be applied to optimize aAPC, and in turn how aAPC platforms can be used to better understand endogenous T cell stimulation. This article is part of a Special Issue entitled: Nanoscale membrane organisation and signalling.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan P Schneck
- Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
86
|
Abstract
The immune system uses much of the classic machinery of cell biology, but in ways that put a different spin on organization and function. Striking recent examples include the demonstration of intraflagellar transport protein and hedgehog contributions to the immune synapse, even though immune cells lack a primary cilium that would be the typical setting for this machinery. In a second example, lymphocytes have their own subfamily of integrins, the β2 subfamily, and only integrins in this family form a stable adhesion ring using freely mobile ligands, a key feature of the immunological synapse. Finally, we showed recently that T-cells use endosomal sorting complexes required for transport (ESCRTs) at the plasma membrane to generate T-cell antigen receptor–enriched microvesicles. It is unusual for the ESCRT pathway to operate at the plasma membrane, but this may allow a novel form of cell–cell communication by providing a multivalent ligand for major histocompatibility complex–peptide complexes and perhaps other receptors on the partnering B-cell. Immune cells are thus an exciting system for novel cell biology even with classical pathways that have been studied extensively in other cell types.
Collapse
Affiliation(s)
- Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington OX3 7FY, United Kingdom; Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
87
|
Hui KL, Balagopalan L, Samelson LE, Upadhyaya A. Cytoskeletal forces during signaling activation in Jurkat T-cells. Mol Biol Cell 2014; 26:685-95. [PMID: 25518938 PMCID: PMC4325839 DOI: 10.1091/mbc.e14-03-0830] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cytoskeletal forces are implicated in T-cell–receptor activation, but their determinants are not known. Traction force microscopy was used to measure forces generated during T-cell activation. Whereas actin dynamics were essential for force generation, myosin contractility played a limited role. T-cells were also found to be mechanosensitive. T-cells are critical for the adaptive immune response in the body. The binding of the T-cell receptor (TCR) with antigen on the surface of antigen-presenting cells leads to cell spreading and signaling activation. The underlying mechanism of signaling activation is not completely understood. Although cytoskeletal forces have been implicated in this process, the contribution of different cytoskeletal components and their spatial organization are unknown. Here we use traction force microscopy to measure the forces exerted by Jurkat T-cells during TCR activation. Perturbation experiments reveal that these forces are largely due to actin assembly and dynamics, with myosin contractility contributing to the development of force but not its maintenance. We find that Jurkat T-cells are mechanosensitive, with cytoskeletal forces and signaling dynamics both sensitive to the stiffness of the substrate. Our results delineate the cytoskeletal contributions to interfacial forces exerted by T-cells during activation.
Collapse
Affiliation(s)
- King Lam Hui
- Department of Physics, University of Maryland, College Park, MD 20742
| | - Lakshmi Balagopalan
- Institute for Physical Sciences and Technology, University of Maryland, College Park, MD 20742
| | - Lawrence E Samelson
- Institute for Physical Sciences and Technology, University of Maryland, College Park, MD 20742
| | - Arpita Upadhyaya
- Department of Physics, University of Maryland, College Park, MD 20742 Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
88
|
Ratzinger F, Haslacher H, Poeppl W, Hoermann G, Kovarik JJ, Jutz S, Steinberger P, Burgmann H, Pickl WF, Schmetterer KG. Azithromycin suppresses CD4(+) T-cell activation by direct modulation of mTOR activity. Sci Rep 2014; 4:7438. [PMID: 25500904 PMCID: PMC4262884 DOI: 10.1038/srep07438] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/21/2014] [Indexed: 12/31/2022] Open
Abstract
Advanced macrolides, such as azithromycin (AZM) or clarithromycin (CLM), are antibiotics with immunomodulatory properties. Here we have sought to evaluate their in vitro influence on the activation of CD4(+) T-cells. Isolated CD4(+) T-cells were stimulated with agonistic anti-CD3/anti-CD28 monoclonal antibodies in the presence of 0.6 mg/L, 2.5 mg/L, 10 mg/L or 40 mg/L AZM or CLM. Cell proliferation, cytokine level in supernatants and cell viability was assessed. Intracellular signaling pathways were evaluated using reporter cell lines, FACS analysis, immunoblotting and in vitro kinase assays. AZM inhibited cell proliferation rate and cytokine secretion of CD4(+) T-cells in a dose-dependent manner. Similarly, high concentrations of CLM (40 mg/L) also suppressed these T-cell functions. Analysis of molecular signaling pathways revealed that exposure to AZM reduced the phosphorylation of the S6 ribosomal protein, a downstream target of mTOR. This effect was also observed at 40 mg/L CLM. In vitro kinase studies using recombinant mTOR showed that AZM inhibited mTOR activity. In contrast to rapamycin, this inhibition was independent of FKBP12. We show for the first time that AZM and to a lesser extent CLM act as immunosuppressive agents on CD4(+) T-cells by inhibiting mTOR activity. Our results might have implications for the clinical use of macrolides.
Collapse
Affiliation(s)
- F. Ratzinger
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - H. Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - W. Poeppl
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Austria
| | - G. Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - J. J. Kovarik
- Clinical Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - S. Jutz
- Institute of Immunology, Medical University of Vienna, Austria
| | - P. Steinberger
- Institute of Immunology, Medical University of Vienna, Austria
| | - H. Burgmann
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Austria
| | - W. F. Pickl
- Institute of Immunology, Medical University of Vienna, Austria
| | - K. G. Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| |
Collapse
|
89
|
Chen B, Jia Y, Gao Y, Sanchez L, Anthony SM, Yu Y. Janus particles as artificial antigen-presenting cells for T cell activation. ACS APPLIED MATERIALS & INTERFACES 2014; 6:18435-9. [PMID: 25343426 PMCID: PMC4404154 DOI: 10.1021/am505510m] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Here we show that the multifunctionality of Janus particles can be exploited for in vitro T cell activation. We engineer bifunctional Janus particles on which the spatial distribution of two ligands, anti-CD3 and fibronectin, mimics the "bull's eye" protein pattern formed in the membrane junction between a T cell and an antigen-presenting cell. Different levels of T cell activation can be achieved by simply switching the spatial distribution of the two ligands on the surfaces of the "bull's eye" particles. We find that the ligand pattern also affects clustering of intracellular proteins. This study demonstrates that anisotropic particles, such as Janus particles, can be developed as artificial antigen-presenting cells for modulating T cell activation.
Collapse
|
90
|
The bullseye synapse formed between CD4+ T-cell and staphylococcal enterotoxin B-pulsed dendritic cell is a suppressive synapse in T-cell response. Immunol Cell Biol 2014; 93:99-110. [PMID: 25287444 DOI: 10.1038/icb.2014.76] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/15/2014] [Accepted: 08/18/2014] [Indexed: 01/05/2023]
Abstract
The immunological synapse (IS) is a supermolecular activation cluster formed between T cells and antigen-presenting cells. Although diverse IS structures have been reported, the function of the IS in T-cell activation remains unclear. Here, we found that the bullseye IS, one of IS types at the interface of CD4(+) T cells and staphylococcal enterotoxin B-pulsed dendritic cells, suppressed CD4(+) T-cell activation, whereas multifocal IS, another synapse type, stimulated CD4(+) T-cell activation. Consistent with these results, bullseye IS formation was accompanied by a low-level calcium response in T cells and a loss of T-cell receptor signalling molecules from the synapse, whereas multifocal IS exhibited the opposite. Furthermore, we found that CD4(+)CD25(+) regulatory T cells (T(regs)) more efficiently formed bullseye IS and promoted bullseye IS formation in CD4(+) CD25(-) T cells. Cytotoxic T-lymphocyte antigen-4 (CTLA-4), an inhibitory molecule expressed continuously on T(regs), was localised in bullseye IS. Moreover, blocking CTLA-4 reduced the percentage of bullseye IS formation and promoted T-cell activation. Our data thus indicate that bullseye IS formation is mediated by CTLA-4, and may negatively control T-cell activation as a suppressive synapse.
Collapse
|
91
|
Kupfer-type immunological synapses in vivo: Raison D'être of SMAC. Immunol Cell Biol 2014; 93:51-6. [PMID: 25267483 DOI: 10.1038/icb.2014.80] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/01/2014] [Accepted: 08/03/2014] [Indexed: 01/04/2023]
Abstract
T cells engage with antigen-presenting cells to form immunological synapses. These intimate contacts are characterized by the complex arrangement of molecules at the intercellular interface, which has been described as the supramolecular activation cluster (SMAC). However, due to T cells functioning without SMAC formation and the difficulties of studying these complex arrangements in vivo, its biological importance has been questioned. In light of recent data, we focus this review on the putative functionality of SMACs in T-cell synaptic contacts in vivo and emphasize the therapeutic potential of SMAC manipulation in immune-driven diseases.
Collapse
|
92
|
Chu C, Wang Y, Zhang X, Ni X, Cao J, Xu W, Dong Z, Yuan P, Wei W, Ma Y, Zhang L, Wu L, Qi H. SAP-regulated T Cell-APC adhesion and ligation-dependent and -independent Ly108-CD3ζ interactions. THE JOURNAL OF IMMUNOLOGY 2014; 193:3860-71. [PMID: 25217164 DOI: 10.4049/jimmunol.1401660] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The germinal center response requires cooperation between Ag-specific T and B lymphocytes, which takes the form of long-lasting cell-cell conjugation in vivo. Signaling lymphocytic activation molecule (SLAM)-associated protein (SAP) is required for stable cognate T-B cell conjugation, whereas SLAM family transmembrane (TM) receptor Ly108 may negatively regulate this process. We show that, other than phosphotyrosine-binding, SAP does not harbor motifs that recruit additional signaling intermediates to stabilize T-B adhesion. Ly108 dampens T cell adhesion to not only Ag-presenting B cells, but also dendritic cells by inhibiting CD3ζ phosphorylation through two levels of regulated Ly108-CD3ζ interactions. Constitutively associated with Src homology 2 domain-containing tyrosine phosphatase-1 even in SAP-competent cells, Ly108 is codistributed with the CD3 complex within a length scale of 100-200 nm on quiescent cells and can reduce CD3ζ phosphorylation in the absence of overt TCR stimulation or Ly108 ligation. When Ly108 is engaged in trans during cell-cell interactions, Ly108-CD3ζ interactions are promoted in a manner that uniquely depends on Ly108 TM domain, leading to more efficient CD3ζ dephosphorylation. Whereas replacement of the Ly108 TM domain still allows the constitutive, colocalization-dependent inhibition of CD3ζ phosphorylation, it abrogates the ligation-dependent Ly108-CD3ζ interactions and CD3ζ dephosphorylation, and it abolishes the suppression on Ag-triggered T-B adhesion. These results offer new insights into how SAP and Ly108 antagonistically modulate the strength of proximal TCR signaling and thereby control cognate T cell-APC interactions.
Collapse
Affiliation(s)
- Coco Chu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Yifeng Wang
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Xu Zhang
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Xinya Ni
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Junxia Cao
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Wan Xu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Zhongjun Dong
- Laboratory of Tumor Immunology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Pengfei Yuan
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, People's Republic of China; and
| | - Wensheng Wei
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, People's Republic of China; and
| | - Yuanwu Ma
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Longyan Wu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China;
| |
Collapse
|
93
|
Roybal KT, Sinai P, Verkade P, Murphy RF, Wülfing C. The actin-driven spatiotemporal organization of T-cell signaling at the system scale. Immunol Rev 2014; 256:133-47. [PMID: 24117818 DOI: 10.1111/imr.12103] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
T cells are activated through interaction with antigen-presenting cells (APCs). During activation, receptors and signaling intermediates accumulate in diverse spatiotemporal distributions. These distributions control the probability of signaling interactions and thus govern information flow through the signaling system. Spatiotemporally resolved system-scale investigation of signaling can extract the regulatory information thus encoded, allowing unique insight into the control of T-cell function. Substantial technical challenges exist, and these are briefly discussed herein. While much of the work assessing T-cell spatiotemporal organization uses planar APC substitutes, we focus here on B-cell APCs with often stark differences. Spatiotemporal signaling distributions are driven by cell biologically distinct structures, a large protein assembly at the interface center, a large invagination, the actin-supported interface periphery as extended by smaller individual lamella, and a newly discovered whole-interface actin-driven lamellum. The more than 60 elements of T-cell activation studied to date are dynamically distributed between these structures, generating a complex organization of the signaling system. Signal initiation and core signaling prefer the interface center, while signal amplification is localized in the transient lamellum. Actin dynamics control signaling distributions through regulation of the underlying structures and drive a highly undulating T-cell/APC interface that imposes substantial constraints on T-cell organization. We suggest that the regulation of actin dynamics, by controlling signaling distributions and membrane topology, is an important rheostat of T-cell signaling.
Collapse
Affiliation(s)
- Kole T Roybal
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | |
Collapse
|
94
|
Beemiller P, Krummel MF. Regulation of T-cell receptor signaling by the actin cytoskeleton and poroelastic cytoplasm. Immunol Rev 2014; 256:148-59. [PMID: 24117819 DOI: 10.1111/imr.12120] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The actin cytoskeleton plays essential roles in modulating T-cell activation. Most models of T-cell receptor (TCR) triggering signalosome assembly and immune synapse formation invoke actin-dependent mechanisms. As T cells are constitutively motile cells, TCR triggering and signaling occur against a cytoskeletal backdrop that is constantly remodeling. While the interplay between actin dynamics and TCR signaling have been the focus of research for many years, much of the work in T cells has considered actin largely for its 'scaffolding' function. We examine the roles of the actin cytoskeleton in TCR signaling and immune synapse formation with an emphasis on how poroelasticity, an ensemble feature of actin dynamics with the cytosol, relates to how T cells respond to stimulation.
Collapse
Affiliation(s)
- Peter Beemiller
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
95
|
Soares H, Lasserre R, Alcover A. Orchestrating cytoskeleton and intracellular vesicle traffic to build functional immunological synapses. Immunol Rev 2014; 256:118-32. [PMID: 24117817 DOI: 10.1111/imr.12110] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunological synapses are specialized cell-cell contacts formed between T lymphocytes and antigen-presenting cells. They are induced upon antigen recognition and are crucial for T-cell activation and effector functions. The generation and function of immunological synapses depend on an active T-cell polarization process, which results from a finely orchestrated crosstalk between the antigen receptor signal transduction machinery, the actin and microtubule cytoskeletons, and controlled vesicle traffic. Although we understand how some of these particular events are regulated, we still lack knowledge on how these multiple cellular elements are harmonized to ensure appropriate T-cell responses. We discuss here our view on how T-cell receptor signal transduction initially commands cytoskeletal and vesicle traffic polarization, which in turn sets the immunological synapse molecular design that regulates T-cell activation. We also discuss how the human immunodeficiency virus (HIV-1) hijacks some of these processes impairing immunological synapse generation and function.
Collapse
Affiliation(s)
- Helena Soares
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, Paris, France; CNRS, URA-1961, Paris, France
| | | | | |
Collapse
|
96
|
González-Granado JM, Silvestre-Roig C, Rocha-Perugini V, Trigueros-Motos L, Cibrián D, Morlino G, Blanco-Berrocal M, Osorio FG, Freije JMP, López-Otín C, Sánchez-Madrid F, Andrés V. Nuclear envelope lamin-A couples actin dynamics with immunological synapse architecture and T cell activation. Sci Signal 2014; 7:ra37. [PMID: 24757177 DOI: 10.1126/scisignal.2004872] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In many cell types, nuclear A-type lamins regulate multiple cellular functions, including higher-order genome organization, DNA replication and repair, gene transcription, and signal transduction; however, their role in specialized immune cells remains largely unexplored. We showed that the abundance of A-type lamins was almost negligible in resting naïve T lymphocytes, but was increased upon activation of the T cell receptor (TCR). The increase in lamin-A was an early event that accelerated formation of the immunological synapse between T cells and antigen-presenting cells. Polymerization of F-actin in T cells is a critical step for immunological synapse formation, and lamin-A interacted with the linker of nucleoskeleton and cytoskeleton (LINC) complex to promote F-actin polymerization. We also showed that lamin-A expression accelerated TCR clustering and led to enhanced downstream signaling, including extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, as well as increased target gene expression. Pharmacological inhibition of the ERK pathway reduced lamin-A-dependent T cell activation. Moreover, mice lacking lamin-A in immune cells exhibited impaired T cell responses in vivo. These findings underscore the importance of A-type lamins for TCR activation and identify lamin-A as a previously unappreciated regulator of the immune response.
Collapse
Affiliation(s)
- José María González-Granado
- Department of Epidemiology, Atherothrombosis and Imaging, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Carlos Silvestre-Roig
- Department of Epidemiology, Atherothrombosis and Imaging, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Vera Rocha-Perugini
- Vascular Biology and Inflammation. CNIC, Madrid, Spain.,Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Laia Trigueros-Motos
- Department of Epidemiology, Atherothrombosis and Imaging, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Danay Cibrián
- Vascular Biology and Inflammation. CNIC, Madrid, Spain.,Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Giulia Morlino
- Vascular Biology and Inflammation. CNIC, Madrid, Spain.,Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Marta Blanco-Berrocal
- Department of Epidemiology, Atherothrombosis and Imaging, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Fernando Garcia Osorio
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo-IUOPA, Oviedo, Spain
| | | | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo-IUOPA, Oviedo, Spain
| | - Francisco Sánchez-Madrid
- Vascular Biology and Inflammation. CNIC, Madrid, Spain.,Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Vicente Andrés
- Department of Epidemiology, Atherothrombosis and Imaging, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
97
|
Fernández-Arenas E, Calleja E, Martínez-Martín N, Gharbi SI, Navajas R, García-Medel N, Penela P, Alcamí A, Mayor F, Albar JP, Alarcón B. β-Arrestin-1 mediates the TCR-triggered re-routing of distal receptors to the immunological synapse by a PKC-mediated mechanism. EMBO J 2014; 33:559-77. [PMID: 24502978 DOI: 10.1002/embj.201386022] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
T-cell receptors (TCR) recognize their antigen ligand at the interface between T cells and antigen-presenting cells, known as the immunological synapse (IS). The IS provides a means of sustaining the TCR signal which requires the continual supply of new TCRs. These are endocytosed and redirected from distal membrane locations to the IS. In our search for novel cytoplasmic effectors, we have identified β-arrestin-1 as a ligand of non-phosphorylated resting TCRs. Using dominant-negative and knockdown approaches we demonstrate that β-arrestin-1 is required for the internalization and downregulation of non-engaged bystander TCRs. Furthermore, TCR triggering provokes the β-arrestin-1-mediated downregulation of the G-protein coupled chemokine receptor CXCR4, but not of other control receptors. We demonstrate that β-arrestin-1 recruitment to the TCR, and bystander TCR and CXCR4 downregulation, are mechanistically mediated by the TCR-triggered PKC-mediated phosphorylation of β-arrestin-1 at Ser163. This mechanism allows the first triggered TCRs to deliver a stop migration signal, and to promote the internalization of distal TCRs and CXCR4 and their translocation to the IS. This receptor crosstalk mechanism is critical to sustain the TCR signal.
Collapse
Affiliation(s)
- Elena Fernández-Arenas
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Sunshine JC, Perica K, Schneck JP, Green JJ. Particle shape dependence of CD8+ T cell activation by artificial antigen presenting cells. Biomaterials 2014; 35:269-277. [PMID: 24099710 PMCID: PMC3902087 DOI: 10.1016/j.biomaterials.2013.09.050] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/14/2013] [Indexed: 12/15/2022]
Abstract
Previous work developing particle-based acellular, artificial antigen presenting cells (aAPCs) has focused exclusively on spherical platforms. To explore the role of shape, we generated ellipsoidal PLGA microparticles with varying aspect ratios (ARs) and synthesized aAPCs from them. The ellipsoidal biomimetic aAPCs with high-AR showed significantly enhanced in vitro and in vivo activity above spherical aAPCs with particle volume and antigen content held constant. Confocal imaging indicates that CD8+ T cells preferentially migrate to and are activated by interaction with the long axis of the aAPC. Importantly, enhanced activity of high-AR aAPCs was seen in a mouse melanoma model, with high-AR aAPCs improving melanoma survival compared to non-cognate aAPCs (p = 0.004) and cognate spherical aAPCs (p = 0.05). These findings indicate that particle geometry is a critical design criterion in the generation of aAPCs, and may offer insight into the essential role of geometry in the interaction between CD8+ T cells and biological APCs.
Collapse
Affiliation(s)
- Joel C. Sunshine
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
| | - Karlo Perica
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
| | - Jonathan P. Schneck
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
| | - Jordan J. Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Department of Ophthalmology Johns Hopkins University School of Medicine, Baltimore, MD. 21231
| |
Collapse
|
99
|
Antigen availability determines CD8⁺ T cell-dendritic cell interaction kinetics and memory fate decisions. Immunity 2013; 39:496-507. [PMID: 24054328 DOI: 10.1016/j.immuni.2013.08.034] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 08/23/2013] [Indexed: 01/29/2023]
Abstract
T cells are activated by antigen (Ag)-bearing dendritic cells (DCs) in lymph nodes in three phases. The duration of the initial phase of transient, serial DC-T cell interactions is inversely correlated with Ag dose. The second phase, characterized by stable DC-T cell contacts, is believed to be necessary for full-fledged T cell activation. Here we have shown that this is not the case. CD8⁺ T cells interacting with DCs presenting low-dose, short-lived Ag did not transition to phase 2, whereas higher Ag dose yielded phase 2 transition. Both antigenic constellations promoted T cell proliferation and effector differentiation but yielded different transcriptome signatures at 12 hr and 24 hr. T cells that experienced phase 2 developed long-lived memory, whereas conditions without stable contacts yielded immunological amnesia. Thus, T cells make fate decisions within hours after Ag exposure, resulting in long-term memory or abortive effector responses, correlating with T cell-DCs interaction kinetics.
Collapse
|
100
|
Chapman NM, Connolly SF, Reinl EL, Houtman JCD. Focal adhesion kinase negatively regulates Lck function downstream of the T cell antigen receptor. THE JOURNAL OF IMMUNOLOGY 2013; 191:6208-21. [PMID: 24227778 DOI: 10.4049/jimmunol.1301587] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Focal adhesion kinase (FAK) is a critical regulator of signal transduction in multiple cell types. Although this protein is activated upon TCR engagement, the cellular function that FAK plays in mature human T cells is unknown. By suppressing the function of FAK, we revealed that FAK inhibits TCR-mediated signaling by recruiting C-terminal Src kinase to the membrane and/or receptor complex following TCR activation. Thus, in the absence of FAK, the inhibitory phosphorylation of Lck and/or Fyn is impaired. Together, these data highlight a novel role for FAK as a negative regulator TCR function in human T cells. These results also suggest that changes in FAK expression could modulate sensitivity to TCR stimulation and contribute to the progression of T cell malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Nicole M Chapman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | | | | | | |
Collapse
|