51
|
Gouttefangeas C, Schuhmacher J, Dimitrov S. Adhering to adhesion: assessing integrin conformation to monitor T cells. Cancer Immunol Immunother 2019; 68:1855-1863. [PMID: 31309255 PMCID: PMC11028104 DOI: 10.1007/s00262-019-02365-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/02/2019] [Indexed: 11/27/2022]
Abstract
Monitoring T cells is of major importance for the development of immunotherapies. Recent sophisticated assays can address particular aspects of the anti-tumor T-cell repertoire or support very large-scale immune screening for biomarker discovery. Robust methods for the routine assessment of the quantity and quality of antigen-specific T cells remain, however, essential. This review discusses selected methods that are commonly used for T-cell monitoring and summarizes the advantages and limitations of these assays. We also present a new functional assay, which specifically detects activated β2 integrins within a very short time following CD8+ T-cell stimulation. Because of its unique and favorable characteristics, this assay could be useful for implementation into our T-cell monitoring toolbox.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany.
| | - Juliane Schuhmacher
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Auf der Morgenstelle 15, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Stoyan Dimitrov
- Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University, Otfried-Müller Straße 25, 72076, Tübingen, Germany.
- German Center for Diabetes Research, 72076, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich at the University of Tübingen (IDM), Otfried-Müller Straße 10, 72076, Tübingen, Germany.
| |
Collapse
|
52
|
Tokunaga A, Sugiyama D, Maeda Y, Warner AB, Panageas KS, Ito S, Togashi Y, Sakai C, Wolchok JD, Nishikawa H. Selective inhibition of low-affinity memory CD8 + T cells by corticosteroids. J Exp Med 2019; 216:2701-2713. [PMID: 31537643 PMCID: PMC6888983 DOI: 10.1084/jem.20190738] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/12/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
Corticosteroids inhibit antitumor immune responses of immune checkpoint blockade in a dose- and timing-dependent manner. Memory CD8+ T cells with low TCR affinity are selectively suppressed by corticosteroids, necessitating careful and thoughtful corticosteroid use. Patients treated with immune checkpoint blockade (ICB) sometimes experience immune-related adverse events (irAEs), requiring immuno-suppressive drugs such as corticosteroids despite the possibility that immunosuppression may impair the antitumor effects of ICB. Here, we address the dilemma of using corticosteroids for the treatment of irAEs induced by ICB. ICB augments neoantigen-specific CD8+ T cell responses, resulting in tumor regression. In our model, simultaneous, but not late, administration of corticosteroids impaired antitumor responses with reduction of CD8+ T cell proliferation. Secondary challenge using tumors with/without the neoantigen showed selective progression in tumors lacking the neoantigen when corticosteroids were administered. Corticosteroids decreased low- but not high-affinity memory T cells by suppressing fatty acid metabolism essential for memory T cells. In a small cohort of human melanoma patients, overall survival was shorter after treatment with CTLA-4 blockade in patients who received early corticosteroids or had low tumor mutation burden. Together, low-affinity memory T cells are dominantly suppressed by corticosteroids, necessitating careful and thoughtful corticosteroid use.
Collapse
Affiliation(s)
- Akihiro Tokunaga
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Oncology Research and Development Unit, Kyowa Kirin Co., Ltd., Shizuoka, Japan
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuka Maeda
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Allison Betof Warner
- Parker Institute for Cancer Immunotherapy, Swim Across America-Ludwig Collaborative Lab, Memorial Sloan-Kettering Cancer Center, New York, NY.,Weill Cornell Medical College, New York, NY
| | - Katherine S Panageas
- Departments of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sachiko Ito
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yosuke Togashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Chika Sakai
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Jedd D Wolchok
- Parker Institute for Cancer Immunotherapy, Swim Across America-Ludwig Collaborative Lab, Memorial Sloan-Kettering Cancer Center, New York, NY .,Weill Cornell Medical College, New York, NY
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan .,Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
53
|
Sivapatham S, Ficht X, Barreto de Albuquerque J, Page N, Merkler D, Stein JV. Initial Viral Inoculum Determines Kinapse-and Synapse-Like T Cell Motility in Reactive Lymph Nodes. Front Immunol 2019; 10:2086. [PMID: 31552034 PMCID: PMC6743022 DOI: 10.3389/fimmu.2019.02086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/19/2019] [Indexed: 11/13/2022] Open
Abstract
T cell activation in lymphoid tissue occurs through interactions with cognate peptide-major histocompatibility complex (pMHC)-presenting dendritic cells (DCs). Intravital imaging studies using ex vivo peptide-pulsed DCs have uncovered that cognate pMHC levels imprint a wide range of dynamic contacts between these two cell types. T cell-DC interactions vary between transient, "kinapse-like" contacts at low to moderate pMHC levels to immediate "synapse-like" arrest at DCs displaying high pMHC levels. To date, it remains unclear whether this pattern is recapitulated when the immune system faces a replicative agent, such as a virus, at low and high inoculum. Here, we locally administered low and high inoculum of lymphocytic choriomeningitis virus (LCMV) in mice to follow activation parameters of Ag-specific CD4+ and CD8+ T cells in draining lymph nodes (LNs) during the first 72 h post infection. We correlated these data with kinapse- and synapse-like motility patterns of Ag-specific T cells obtained by intravital imaging of draining LNs. Our data show that initial viral inoculum controls immediate synapse-like T cell arrest vs. continuous kinapse-like motility. This remains the case when the viral inoculum and thus the inflammatory microenvironment in draining LNs remains identical but cognate pMHC levels vary. Our data imply that the Ag-processing capacity of draining LNs is equipped to rapidly present high levels of cognate pMHC when antigenic material is abundant. Our findings further suggest that widespread T cell arrest during the first 72 h of an antimicrobial immune responses is not required to trigger proliferation. In sum, T cells adapt their scanning behavior according to available antigen levels during viral infections, with dynamic changes in motility occurring before detectable expression of early activation markers.
Collapse
Affiliation(s)
- Sujana Sivapatham
- Department of Oncology, Microbiology, and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Xenia Ficht
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | - Nicolas Page
- Division of Clinical Pathology, Department of Pathology and Immunology, University Hospital of Geneva, Geneva, Switzerland
| | - Doron Merkler
- Division of Clinical Pathology, Department of Pathology and Immunology, University Hospital of Geneva, Geneva, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology, and Immunology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
54
|
Ugur M, Mueller SN. T cell and dendritic cell interactions in lymphoid organs: More than just being in the right place at the right time. Immunol Rev 2019; 289:115-128. [DOI: 10.1111/imr.12753] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/31/2019] [Accepted: 02/03/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Milas Ugur
- Department of Microbiology and Immunology The University of Melbourne, The Peter Doherty Institute for Infection and Immunity Melbourne Victoria Australia
| | - Scott N. Mueller
- Department of Microbiology and Immunology The University of Melbourne, The Peter Doherty Institute for Infection and Immunity Melbourne Victoria Australia
- The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne Melbourne Victoria Australia
| |
Collapse
|
55
|
Dimitrov S, Lange T, Gouttefangeas C, Jensen ATR, Szczepanski M, Lehnnolz J, Soekadar S, Rammensee HG, Born J, Besedovsky L. Gα s-coupled receptor signaling and sleep regulate integrin activation of human antigen-specific T cells. J Exp Med 2019; 216:517-526. [PMID: 30755455 PMCID: PMC6400544 DOI: 10.1084/jem.20181169] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 12/03/2018] [Accepted: 01/11/2019] [Indexed: 12/26/2022] Open
Abstract
This study demonstrates a regulatory role of Gαs-coupled receptor agonists (catecholamines, prostaglandins, and adenosine) and sleep on integrin activation on T cells in humans. The findings point to a mechanism by which T cell responses are altered in several conditions characterized by aberrant levels of these substances. Efficient T cell responses require the firm adhesion of T cells to their targets, e.g., virus-infected cells, which depends on T cell receptor (TCR)–mediated activation of β2-integrins. Gαs-coupled receptor agonists are known to have immunosuppressive effects, but their impact on TCR-mediated integrin activation is unknown. Using multimers of peptide major histocompatibility complex molecules (pMHC) and of ICAM-1—the ligand of β2-integrins—we show that the Gαs-coupled receptor agonists isoproterenol, epinephrine, norepinephrine, prostaglandin (PG) E2, PGD2, and adenosine strongly inhibit integrin activation on human CMV- and EBV-specific CD8+ T cells in a dose-dependent manner. In contrast, sleep, a natural condition of low levels of Gαs-coupled receptor agonists, up-regulates integrin activation compared with nocturnal wakefulness, a mechanism possibly underlying some of the immune-supportive effects of sleep. The findings are also relevant for several pathologies associated with increased levels of Gαs-coupled receptor agonists (e.g., tumor growth, malaria, hypoxia, stress, and sleep disturbances).
Collapse
Affiliation(s)
- Stoyan Dimitrov
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany .,German Center for Diabetes Research (DZD), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Tanja Lange
- Clinic for Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Anja T R Jensen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Szczepanski
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Jannik Lehnnolz
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Surjo Soekadar
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany.,Clinical Neurotechnology Laboratory, Department of Psychiatry and Psychotherapy, Neuroscience Research Center, Charité - University Medicine Berlin, Berlin, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany.,Partner Site Tübingen, German Cancer Consortium, Tübingen, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Luciana Besedovsky
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
56
|
Yan L, Liu B. Critical factors in chimeric antigen receptor-modified T-cell (CAR-T) therapy for solid tumors. Onco Targets Ther 2018; 12:193-204. [PMID: 30636882 PMCID: PMC6309774 DOI: 10.2147/ott.s190336] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The success of chimeric antigen receptor-modified T-cell (CAR-T) therapy for B-cell lymphocyte malignancies targeting CD19 places it in a rapidly growing field in cancer immunotherapy for both hematological and solid tumors. However, the two types of tumor are quite different in the following respects. Solid tumors are characterized by complex vasculatures and matrix barriers that significantly affect T-cell functions and migration. Moreover, various immunosuppressive molecules expressed in the tumor microenvironment can impede T-cell activation, and the high metabolic rate of tumors competitively suppresses the metabolism of immune cells. All these factors will exert their influences on the development of a cancer, which is a dynamic balance between the host's immune system and the tumor. At present, solid tumors are treated primarily by surgical resection combined with radiotherapy and chemotherapy, a treatment process that is painful and not always effective. With advantages over traditional treatments, the recently developed CAR-T immunotherapy has been applied and has shown highly promising results. Nevertheless, the complexity of solid tumors presents a great challenge to this technique. This review focuses on elucidating the factors influencing the anti-tumor effects of CAR-T in the specific tumor environment, and hence exploring feasible approaches to overcome them.
Collapse
Affiliation(s)
- Lingli Yan
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou, China,
| | - Bainan Liu
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou, China,
| |
Collapse
|
57
|
Xia F, Qian CR, Xun Z, Hamon Y, Sartre AM, Formisano A, Mailfert S, Phelipot MC, Billaudeau C, Jaeger S, Nunès JA, Guo XJ, He HT. TCR and CD28 Concomitant Stimulation Elicits a Distinctive Calcium Response in Naive T Cells. Front Immunol 2018; 9:2864. [PMID: 30564247 PMCID: PMC6288997 DOI: 10.3389/fimmu.2018.02864] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/20/2018] [Indexed: 11/13/2022] Open
Abstract
T cell activation is initiated upon ligand engagement of the T cell receptor (TCR) and costimulatory receptors. The CD28 molecule acts as a major costimulatory receptor in promoting full activation of naive T cells. However, despite extensive studies, why naive T cell activation requires concurrent stimulation of both the TCR and costimulatory receptors remains poorly understood. Here, we explore this issue by analyzing calcium response as a key early signaling event to elicit T cell activation. Experiments using mouse naive CD4+ T cells showed that engagement of the TCR or CD28 with the respective cognate ligand was able to trigger a rise in fluctuating calcium mobilization levels, as shown by the frequency and average response magnitude of the reacting cells compared with basal levels occurred in unstimulated cells. The engagement of both TCR and CD28 enabled a further increase of these two metrics. However, such increases did not sufficiently explain the importance of the CD28 pathways to the functionally relevant calcium responses in T cell activation. Through the autocorrelation analysis of calcium time series data, we found that combined but not separate TCR and CD28 stimulation significantly prolonged the average decay time (τ) of the calcium signal amplitudes determined with the autocorrelation function, compared with its value in unstimulated cells. This increasement of decay time (τ) uniquely characterizes the fluctuating calcium response triggered by concurrent stimulation of TCR and CD28, as it could not be achieved with either stronger TCR stimuli or by co-engaging both TCR and LFA-1, and likely represents an important feature of competent early signaling to provoke efficient T cell activation. Our work has thus provided new insights into the interplay between the TCR and CD28 early signaling pathways critical to trigger naive T cell activation.
Collapse
Affiliation(s)
- Fan Xia
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Cheng-Rui Qian
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Zhou Xun
- School of Economics, Jiangxi University of Finance and Economics, Nanchang, China.,Aix Marseille University, AMSE and GREQAM, Marseille, France
| | - Yannick Hamon
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | | | | | | | | | | | | | - Jacques A Nunès
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.,Equipe Labellisée Fondation pour la Recherche Médicale, Centre de Recherche en Cancérologie de Marseille, Immunology and Cancer, Marseille, France
| | - Xiao-Jun Guo
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Hai-Tao He
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| |
Collapse
|
58
|
Polarity of CD4+ T cells towards the antigen presenting cell is regulated by the Lck adapter TSAd. Sci Rep 2018; 8:13319. [PMID: 30190583 PMCID: PMC6127336 DOI: 10.1038/s41598-018-31510-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/08/2018] [Indexed: 01/02/2023] Open
Abstract
Polarization of T cells towards the antigen presenting cell (APC) is critically important for appropriate activation and differentiation of the naïve T cell. Here we used imaging flow cytometry (IFC) and show that the activation induced Lck and Itk adapter T cell specific adapter protein (TSAd), encoded by SH2D2A, modulates polarization of T cells towards the APC. Upon exposure to APC presenting the cognate antigen Id, Sh2d2a−/− CD4+ T cells expressing Id-specific transgenic T cell receptor (TCR), displayed impaired polarization of F-actin and TCR to the immunological synapse (IS). Sh2d2a−/− T-cells that did polarize F-actin and TCR still displayed impaired polarization of PKCξ, PAR3 and the microtubule-organizing center (MTOC). In vitro differentiation of activated Sh2d2a−/− T cells was skewed towards an effector memory (Tem) rather than a central memory (Tcm) phenotype. A similar trend was observed for Id-specific TCR Sh2d2a−/− T cells stimulated with APC and cognate antigen. Taken together our data suggest that TSAd modulates differentiation of experienced T cells possibly through polarization of CD4+ T cells towards the APC.
Collapse
|
59
|
Wang J, Lin F, Wan Z, Sun X, Lu Y, Huang J, Wang F, Zeng Y, Chen YH, Shi Y, Zheng W, Li Z, Xiong C, Liu W. Profiling the origin, dynamics, and function of traction force in B cell activation. Sci Signal 2018; 11:11/542/eaai9192. [PMID: 30087179 DOI: 10.1126/scisignal.aai9192] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
B lymphocytes use B cell receptors (BCRs) to recognize membrane-bound antigens to further initiate cell spreading and contraction responses during B cell activation. We combined traction force microscopy and live-cell imaging to profile the origin, dynamics, and function of traction force generation in these responses. We showed that B cell activation required the generation of 10 to 20 nN of traction force when encountering antigens presented by substrates with stiffness values from 0.5 to 1 kPa, which mimic the rigidity of antigen-presenting cells in vivo. Perturbation experiments revealed that F-actin remodeling and myosin- and dynein-mediated contractility contributed to traction force generation and B cell activation. Moreover, membrane-proximal BCR signaling molecules (including Lyn, Syk, Btk, PLC-γ2, BLNK, and Vav3) and adaptor molecules (Grb2, Cbl, and Dok-3) linking BCR microclusters and motor proteins were also required for the sustained generation of these traction forces. We found a positive correlation between the strength of the traction force and the mean fluorescence intensity of the BCR microclusters. Furthermore, we demonstrated that isotype-switched memory B cells expressing immunoglobulin G (IgG)-BCRs generated greater traction forces than did mature naïve B cells expressing IgM-BCRs during B cell activation. Last, we observed that primary B cells from patients with rheumatoid arthritis generated greater traction forces than did B cells from healthy donors in response to antigen stimulation. Together, these data delineate the origin, dynamics, and function of traction force during B cell activation.
Collapse
Affiliation(s)
- Junyi Wang
- China Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Feng Lin
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Zhengpeng Wan
- China Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China
| | - Yun Lu
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing 100084, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Fei Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, No. 9 Section 4, Renmin South Road, Chengdu 610041, China
| | - Yingyue Zeng
- School of Life Science, Liaoning University, Shenyang 110036, China
| | - Ying-Hua Chen
- China Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Yan Shi
- Center for Life Sciences, Department of Basic Medical Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Wenjie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China. .,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Wanli Liu
- China Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China. .,Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China
| |
Collapse
|
60
|
Lei AH, Xiao Q, Liu GY, Shi K, Yang Q, Li X, Liu YF, Wang HK, Cai WP, Guan YJ, Gabrilovich DI, Zhou J. ICAM-1 controls development and function of ILC2. J Exp Med 2018; 215:2157-2174. [PMID: 30049704 PMCID: PMC6080904 DOI: 10.1084/jem.20172359] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 05/02/2018] [Accepted: 07/03/2018] [Indexed: 12/12/2022] Open
Abstract
ILC2s are key players in allergic airway inflammation. Lei et al. show that ICAM-1 controls ILC2 development and function through regulating ERK signaling pathway, suggesting targeting ICAM-1 as a potential strategy for ILC2-induced asthma. Group 2 innate lymphoid cells (ILC2s) are emerging as key players in the pathogenesis of allergic airway inflammation. The mechanisms regulating ILC2, however, are not fully understood. Here, we found that ICAM-1 is required for the development and function of ILC2. ICAM-1–deficient (ICAM-1−/−) mice displayed significantly lower levels of ILC2s in the bone marrow and peripheral tissues than wild-type controls. CLP transfer and in vitro culture assays revealed that the regulation of ILC2 by ICAM-1 is cell intrinsic. Furthermore, ILC2s from ICAM-1−/− mice were functionally impaired, as indicated by the diminished production of type-2 cytokines in response to IL-33 challenge. The reduction in lung ILC2s caused a clear remission of airway inflammation in ICAM-1−/− mice after administration of papain or Alternaria alternata. We further demonstrate that ILC2 defects caused by ICAM-1 deficiency are due to ERK signaling-dependent down-regulation of GATA3 protein. Collectively, these observations identify ICAM-1 as a novel regulator of ILC2.
Collapse
Affiliation(s)
- Ai-Hua Lei
- Joint Program in Immunology, Department of Internal Medicine, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiang Xiao
- Joint Program in Immunology, Department of Internal Medicine, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Gao-Yu Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kun Shi
- Department of Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Qiong Yang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xing Li
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu-Feng Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hai-Kun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | | | | | - Dmitry I Gabrilovich
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,The Wistar Institute, Philadelphia, PA.,Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jie Zhou
- Joint Program in Immunology, Department of Internal Medicine, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China .,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
61
|
Liu YF, Zhang SY, Chen YY, Shi K, Zou B, Liu J, Yang Q, Jiang H, Wei L, Li CZ, Zhao M, Gabrilovich DI, Zhang H, Zhou J. ICAM-1 Deficiency in the Bone Marrow Niche Impairs Quiescence and Repopulation of Hematopoietic Stem Cells. Stem Cell Reports 2018; 11:258-273. [PMID: 29937143 PMCID: PMC6117479 DOI: 10.1016/j.stemcr.2018.05.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 12/31/2022] Open
Abstract
The bone marrow niche plays a critical role in controlling the fate of hematopoietic stem cells (HSCs) by integrating intrinsic and extrinsic signals. However, the molecular events in the HSC niche remain to be investigated. Here, we report that intercellular adhesion molecule-1 (ICAM-1) maintains HSC quiescence and repopulation capacity in the niche. ICAM-1-deficient mice (ICAM-1−/−) displayed significant expansion of phenotypic long-term HSCs with impaired quiescence, as well as favoring myeloid cell expansion. ICAM-1-deficient HSCs presented normal reconstitution capacity during serial transplantation; however, reciprocal transplantation experiments showed that ICAM-1 deficiency in the niche impaired HSC quiescence and repopulation capacity. In addition, ICAM-1 deletion caused failure to retain HSCs in the bone marrow and changed the expression profile of stroma cell-derived factors, possibly representing the mechanism for defective HSCs in ICAM-1−/− mice. Collectively, these observations identify ICAM-1 as a regulator in the bone marrow niche. ICAM-1 deficiency expands HSC−LT with impaired quiescence and repopulation The defects characterizing HSC−LT in ICAM-1−/− mice are niche cell dependent ICAM-1−/− niche brings about impaired bone marrow retention and homing of HSC−LT ICAM-1 in human stroma cells might affect the progression of myelocytic leukemia
Collapse
Affiliation(s)
- Yu-Feng Liu
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Shao-Ying Zhang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xian 710000, China; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying-Ying Chen
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Kun Shi
- Guangzhou Women and Children's Medical Center, Guangzhou 510000, China
| | - Bin Zou
- Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Jun Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Qiong Yang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hua Jiang
- Guangzhou Women and Children's Medical Center, Guangzhou 510000, China
| | - Lai Wei
- Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Chang-Zheng Li
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Guangzhou 510080, China
| | - Meng Zhao
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Guangzhou 510080, China
| | - Dmitry I Gabrilovich
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; The Wistar Institute, Philadelphia, PA 19104, USA
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China.
| | - Jie Zhou
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
62
|
Strominger I, Elyahu Y, Berner O, Reckhow J, Mittal K, Nemirovsky A, Monsonego A. The Choroid Plexus Functions as a Niche for T-Cell Stimulation Within the Central Nervous System. Front Immunol 2018; 9:1066. [PMID: 29868025 PMCID: PMC5962702 DOI: 10.3389/fimmu.2018.01066] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 04/27/2018] [Indexed: 12/20/2022] Open
Abstract
The choroid plexus (CP) compartment in the ventricles of the brain comprises fenestrated vasculature and, therefore, it is permeable to blood-borne mediators of inflammation. Here, we explored whether T-cell activation in the CP plays a role in regulating central nervous system (CNS) inflammation. We show that CD4 T cells injected into the lateral ventricles adhere to the CP, transmigrate across its epithelium, and undergo antigen-specific activation and proliferation. This process is enhanced following peripheral immune stimulation and significantly impacts the immune signaling induced by the CP. Ex vivo studies demonstrate that T-cell harboring the CP through its apical surface is a chemokine- and adhesion molecule-dependent process. We suggest that, within the CNS, the CP serves an immunological niche, which rapidly responds to peripheral inflammation and, thereby, promotes two-way T-cell trafficking that impact adaptive immunity in the CNS.
Collapse
Affiliation(s)
- Itai Strominger
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yehezqel Elyahu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omer Berner
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jensen Reckhow
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kritika Mittal
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Anna Nemirovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Alon Monsonego
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
63
|
Bohineust A, Garcia Z, Beuneu H, Lemaître F, Bousso P. Termination of T cell priming relies on a phase of unresponsiveness promoting disengagement from APCs and T cell division. J Exp Med 2018; 215:1481-1492. [PMID: 29588347 PMCID: PMC5940264 DOI: 10.1084/jem.20171708] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/31/2018] [Accepted: 03/07/2018] [Indexed: 01/28/2023] Open
Abstract
Bohineust et al. establish that recently activated T cells exhibit a phase of unresponsiveness associated with a defect in calcium entry. This stage was essential to terminate priming, distracting T cells from APCs, and favoring their clonal expansion. T cells are primed in secondary lymphoid organs by establishing stable interactions with antigen-presenting cells (APCs). However, the cellular mechanisms underlying the termination of T cell priming and the initiation of clonal expansion remain largely unknown. Using intravital imaging, we observed that T cells typically divide without being associated to APCs. Supporting these findings, we demonstrate that recently activated T cells have an intrinsic defect in establishing stable contacts with APCs, a feature that was reflected by a blunted capacity to stop upon T cell receptor (TCR) engagement. T cell unresponsiveness was caused, in part, by a general block in extracellular calcium entry. Forcing TCR signals in activated T cells antagonized cell division, suggesting that T cell hyporesponsiveness acts as a safeguard mechanism against signals detrimental to mitosis. We propose that transient unresponsiveness represents an essential phase of T cell priming that promotes T cell disengagement from APCs and favors effective clonal expansion.
Collapse
Affiliation(s)
- Armelle Bohineust
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Medicale, U1223, Paris, France
| | - Zacarias Garcia
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Medicale, U1223, Paris, France
| | - Hélène Beuneu
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Medicale, U1223, Paris, France
| | - Fabrice Lemaître
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Medicale, U1223, Paris, France
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, Paris, France .,Institut National de la Santé et de la Recherche Medicale, U1223, Paris, France
| |
Collapse
|
64
|
Imaging of cytotoxic antiviral immunity while considering the 3R principle of animal research. J Mol Med (Berl) 2018; 96:349-360. [PMID: 29460050 DOI: 10.1007/s00109-018-1628-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/01/2018] [Accepted: 02/05/2018] [Indexed: 10/18/2022]
Abstract
Adoptive cell transfer approaches for antigen-specific CD8+ T cells are used widely to study their effector potential during infections or cancer. However, contemporary methodological adaptations regarding transferred cell numbers, advanced imaging, and the 3R principle of animal research have been largely omitted. Here, we introduce an improved cell transfer method that reduces the number of donor animals substantially and fulfills the requirements for intravital imaging under physiological conditions. For this, we analyzed the well-established Friend retrovirus (FV) mouse model. Donor mice that expressed a FV-specific T cell receptor (TCRtg) and the fluorescent protein tdTomato were used as source of antigen-specific CD8+ T cells. Only a few drops of peripheral blood were sufficient to isolate ~ 150,000 naive reporter cells from which 1000 were adoptively transferred into recently FV-infected recipients. The cells became activated and functional and expanded strongly in the spleen and bone marrow within 10 days post infection. Transferred CD8+ T cells participated in the antiviral host response within a natural range and developed an effector phenotype indistinguishable from endogenous effector CD8+ T cells. Additionally, the generated reporter cell frequency allowed single cell visualization and tracking of a physiological antiretroviral CD8+ T cell response by intravital two-photon microscopy. Highly reproducible results were obtained in independent experiments by reusing the same donors repetitively for multiple transfers. Our approach allows a strong reduction of experimental animals required for studies on antigen-specific CD8+ T cell function and should be applicable to other transfer models. KEY MESSAGES TCRtg CD8+ T cells are obtained repetitively from the blood samples of single donors. One thousand transferred TCRtg CD8+ T cells get activated, are functional, and proliferate. Several adoptive cell transfers from the same donor show reproducible results. One thousand transferred cells take part in the FV immune response without modifying it. Use of fluorescent transfer cells allows in vivo imaging and single cell tracking.
Collapse
|
65
|
Bertoni A, Alabiso O, Galetto AS, Baldanzi G. Integrins in T Cell Physiology. Int J Mol Sci 2018; 19:E485. [PMID: 29415483 PMCID: PMC5855707 DOI: 10.3390/ijms19020485] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 01/19/2018] [Accepted: 02/02/2018] [Indexed: 11/16/2022] Open
Abstract
From the thymus to the peripheral lymph nodes, integrin-mediated interactions with neighbor cells and the extracellular matrix tune T cell behavior by organizing cytoskeletal remodeling and modulating receptor signaling. LFA-1 (αLβ2 integrin) and VLA-4 (α4β1 integrin) play a key role throughout the T cell lifecycle from thymocyte differentiation to lymphocyte extravasation and finally play a fundamental role in organizing immune synapse, providing an essential costimulatory signal for the T cell receptor. Apart from tuning T cell signaling, integrins also contribute to homing to specific target organs as exemplified by the importance of α4β7 in maintaining the gut immune system. However, apart from those well-characterized examples, the physiological significance of the other integrin dimers expressed by T cells is far less understood. Thus, integrin-mediated cell-to-cell and cell-to-matrix interactions during the T cell lifespan still represent an open field of research.
Collapse
Affiliation(s)
- Alessandra Bertoni
- Department of Translational Medicine and Institute for Research and Cure of Autoimmune Diseases, University of Piemonte Orientale, 28100 Novara, Italy.
| | - Oscar Alabiso
- Department of Translational Medicine, University of Eastern Piedmont, Novara-Italy and Oncology Division, University Hospital "Maggiore della Carità", 28100 Novara, Italy.
| | - Alessandra Silvia Galetto
- Department of Translational Medicine, University of Eastern Piedmont, Novara 28100-Italy and Palliative Care Division, A.S.L., 13100 Vercelli, Italy.
| | - Gianluca Baldanzi
- Department of Translational Medicine and Institute for Research and Cure of Autoimmune Diseases, University of Piemonte Orientale, 28100 Novara, Italy.
| |
Collapse
|
66
|
Monosodium urate crystals induced ICAM-1 expression and cell-cell adhesion in renal mesangial cells: Implications for the pathogenesis of gouty nephropathy. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2018; 53:23-32. [PMID: 29657028 DOI: 10.1016/j.jmii.2017.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 12/11/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Renal disease is prevalent in gouty patients and monosodium urate (MSU) crystal deposition in the kidney can be detected in some gouty nephropathy patients. MSU crystals can induce inflammatory events, we investigated the MSU-induced expression of intercellular adhesion molecule (ICAM)-1 on human renal mesangial cells (HRMCs) and the involved signal transduction mechanisms. METHODS The HRMCs cell line was purchased from ScienCell Research Laboratories. MSU crystals were made by dissolving uric acid in sodium hydroxide (NaOH) solution. The involvement of MAPKs, apoptosis-associated speck-like protein containing a CARD domain (ASC), and Toll-like receptor (TLR) was investigated using pharmacological inhibitors, transfection with short hairpin RNA (shRNA), or monoclonal antibodies. Protein expression was evaluated by Western blotting. The functional activity of ICAM-1 was evaluated with cell-cell adhesion assay and immunofluorescence analysis. RESULTS MSU stimulation increased expression of ICAM-1 and adhesion between HRMCs and human monocytic THP-1 cells. The interaction between HRMCs and THP-1 was suppressed by ICAM-1 neutralizing antibodies. MSU stimulation induced activation of mitogen-activated protein kinases, including c-Jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK), but only p38 was responsible for MSU-induced expression of ICAM-1 and cell-cell adhesion. ASC also play a role in MSU-induced effects. Pretreatment with monoclonal antibodies against toll-like receptor (TLR)2 or TLR4 reduced MSU-induced ICAM-1 expression, cell-cell adhesion, p38 phosphorylation but the reduction of ASC activation is insignificant. CONCLUSION The MSU induced ICAM-1 expression on HRMCs and cell-cell adhesion involved TLR2/4-p38-ICAM1 pathway and TLR2/4 independent ASC-p38-ICAM1 axis. These findings might partly explain the mechanisms underlying gouty nephropathy.
Collapse
|
67
|
Takahashi T, Hsiao HM, Tanaka S, Li W, Higashikubo R, Scozzi D, Bharat A, Ritter JH, Krupnick AS, Gelman AE, Kreisel D. PD-1 expression on CD8 + T cells regulates their differentiation within lung allografts and is critical for tolerance induction. Am J Transplant 2018; 18:216-225. [PMID: 28730633 PMCID: PMC5739961 DOI: 10.1111/ajt.14437] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/18/2017] [Accepted: 07/11/2017] [Indexed: 01/25/2023]
Abstract
Immunological requirements for rejection and tolerance induction differ between various organs. While memory CD8+ T cells are considered a barrier to immunosuppression-mediated acceptance of most tissues and organs, tolerance induction after lung transplantation is critically dependent on central memory CD8+ T lymphocytes. Here we demonstrate that costimulation blockade-mediated tolerance after lung transplantation is dependent on programmed cell death 1 (PD-1) expression on CD8+ T cells. In the absence of PD-1 expression, CD8+ T cells form prolonged interactions with graft-infiltrating CD11c+ cells; their differentiation is skewed towards an effector memory phenotype and grafts are rejected acutely. These findings extend the notion that requirements for tolerance induction after lung transplantation differ from other organs. Thus, immunosuppressive strategies for lung transplant recipients need to be tailored based on the unique immunological properties of this organ.
Collapse
Affiliation(s)
- T Takahashi
- Department of Surgery, Washington University in St. Louis
| | - HM Hsiao
- Department of Surgery, Washington University in St. Louis
| | - S Tanaka
- Department of Surgery, Washington University in St. Louis
| | - W Li
- Department of Surgery, Washington University in St. Louis
| | - R Higashikubo
- Department of Surgery, Washington University in St. Louis
| | - D Scozzi
- Department of Surgery, Washington University in St. Louis
| | - A Bharat
- Department of Surgery, Northwestern University
| | - JH Ritter
- Department of Pathology & Immunology, Washington University in St. Louis
| | - AS Krupnick
- Department of Surgery, University of Virginia
| | - AE Gelman
- Department of Surgery, Washington University in St. Louis,Department of Pathology & Immunology, Washington University in St. Louis
| | - D Kreisel
- Department of Surgery, Washington University in St. Louis,Department of Pathology & Immunology, Washington University in St. Louis,Corresponding author: Daniel Kreisel, M.D., Ph.D., Professor of Surgery, Pathology & Immunology, Campus Box 8234, 660 South Euclid Avenue, Washington University School of Medicine, St. Louis, MO 63110, Tel: (314) 362-6021, Fax: (314) 367-8459,
| |
Collapse
|
68
|
Feigelson SW, Solomon A, Biram A, Hatzav M, Lichtenstein M, Regev O, Kozlovski S, Varol D, Curato C, Leshkowitz D, Jung S, Shulman Z, Alon R. ICAMs Are Not Obligatory for Functional Immune Synapses between Naive CD4 T Cells and Lymph Node DCs. Cell Rep 2018; 22:849-859. [DOI: 10.1016/j.celrep.2017.12.103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/12/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022] Open
|
69
|
Capece T, Walling BL, Lim K, Kim KD, Bae S, Chung HL, Topham DJ, Kim M. A novel intracellular pool of LFA-1 is critical for asymmetric CD8 + T cell activation and differentiation. J Cell Biol 2017; 216:3817-3829. [PMID: 28954823 PMCID: PMC5674876 DOI: 10.1083/jcb.201609072] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/13/2017] [Accepted: 07/11/2017] [Indexed: 11/22/2022] Open
Abstract
The integrin lymphocyte function-associated antigen 1 (LFA-1; CD11a/CD18) is a key T cell adhesion receptor that mediates stable interactions with antigen-presenting cell (APC), as well as chemokine-mediated migration. Using our newly generated CD11a-mYFP knock-in mice, we discovered that naive CD8+ T cells reserve a significant intracellular pool of LFA-1 in the uropod during migration. Intracellular LFA-1 quickly translocated to the cell surface with antigenic stimulus. Importantly, the redistribution of intracellular LFA-1 at the contact with APC was maintained during cell division and led to an unequal inheritance of LFA-1 in divided T cells. The daughter CD8+ T cells with disparate LFA-1 expression showed different patterns of migration on ICAM-1, APC interactions, and tissue retention, as well as altered effector functions. In addition, we identified Rab27 as an important regulator of the intracellular LFA-1 translocation. Collectively, our data demonstrate that an intracellular pool of LFA-1 in naive CD8+ T cells plays a key role in T cell activation and differentiation.
Collapse
Affiliation(s)
- Tara Capece
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY
| | - Brandon L Walling
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY
| | - Kihong Lim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY
| | - Kyun-Do Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY
| | - Seyeon Bae
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY
| | - Hung-Li Chung
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY
| | - David J Topham
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY
| |
Collapse
|
70
|
Zaretsky I, Atrakchi O, Mazor RD, Stoler-Barak L, Biram A, Feigelson SW, Gitlin AD, Engelhardt B, Shulman Z. ICAMs support B cell interactions with T follicular helper cells and promote clonal selection. J Exp Med 2017; 214:3435-3448. [PMID: 28939548 PMCID: PMC5679169 DOI: 10.1084/jem.20171129] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/07/2017] [Accepted: 08/28/2017] [Indexed: 01/13/2023] Open
Abstract
The molecular mechanism governing affinity-based B cell selection for germinal center colonization is unclear. Zaretsky et al. show that B cell ICAMs promote efficient B cell selection for clonal expansion by supporting sustained interactions with T follicular helper cells. The germinal center (GC) reaction begins with a diverse and expanded group of B cell clones bearing a wide range of antibody affinities. During GC colonization, B cells engage in long-lasting interactions with T follicular helper (Tfh) cells, a process that depends on antigen uptake and antigen presentation to the Tfh cells. How long-lasting T–B interactions and B cell clonal expansion are regulated by antigen presentation remains unclear. Here, we use in vivo B cell competition models and intravital imaging to examine the adhesive mechanisms governing B cell selection for GC colonization. We find that intercellular adhesion molecule 1 (ICAM-1) and ICAM-2 on B cells are essential for long-lasting cognate Tfh–B cell interactions and efficient selection of low-affinity B cell clones for proliferative clonal expansion. Thus, B cell ICAMs promote efficient antibody immune response by enhancement of T cell help to cognate B cells.
Collapse
Affiliation(s)
- Irina Zaretsky
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ofir Atrakchi
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Roei D Mazor
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Liat Stoler-Barak
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Adi Biram
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Sara W Feigelson
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Alexander D Gitlin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | | | - Ziv Shulman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
71
|
Abstract
Myeloid cells make extensive use of the complement system in the context of recruitment, phagocytosis, and other effector functions. There are several types of complement receptors on myeloid cells, including G protein-coupled receptors for localizing the source of complement activation, and three sets of type I transmembrane proteins that link complement to phagocytosis: complement receptor 1, having an extracellular domain with tandem complement regulatory repeats; complement receptors 3 and 4, which are integrin family receptors comprising heterodimers of type I transmembrane subunits; and VSIG4, a member of the Ig superfamily. This review will focus on the role of the different classes of complement receptors and how their activities are integrated in the setting of immune tolerance and inflammatory responses.
Collapse
|
72
|
Ackerknecht M, Gollmer K, Germann P, Ficht X, Abe J, Fukui Y, Swoger J, Ripoll J, Sharpe J, Stein JV. Antigen Availability and DOCK2-Driven Motility Govern CD4+ T Cell Interactions with Dendritic Cells In Vivo. THE JOURNAL OF IMMUNOLOGY 2017; 199:520-530. [DOI: 10.4049/jimmunol.1601148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 05/09/2017] [Indexed: 01/07/2023]
|
73
|
NDR1-Dependent Regulation of Kindlin-3 Controls High-Affinity LFA-1 Binding and Immune Synapse Organization. Mol Cell Biol 2017; 37:MCB.00424-16. [PMID: 28137909 PMCID: PMC5376635 DOI: 10.1128/mcb.00424-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/24/2017] [Indexed: 12/23/2022] Open
Abstract
Antigen-specific adhesion between T cells and antigen-presenting cells (APC) during the formation of the immunological synapse (IS) is mediated by LFA-1 and ICAM-1. Here, LFA-1–ICAM-1 interactions were measured at the single-molecule level on supported lipid bilayers. High-affinity binding was detected at low frequencies in the inner peripheral supramolecular activation cluster (SMAC) zone that contained high levels of activated Rap1 and kindlin-3. Rap1 was essential for T cell attachment, whereas deficiencies of ste20-like kinases, Mst1/Mst2, diminished high-affinity binding and abrogated central SMAC (cSMAC) formation with mislocalized kindlin-3 and vesicle transport regulators involved in T cell receptor recycling/releasing machineries, resulting in impaired T cell-APC interactions. We found that NDR1 kinase, activated by the Rap1 signaling cascade through RAPL and Mst1/Mst2, associated with and recruited kindlin-3 to the IS, which was required for high-affinity LFA-1/ICAM-1 binding and cSMAC formation. Our findings reveal crucial roles for Rap1 signaling via NDR1 for recruitment of kindlin-3 and IS organization.
Collapse
|
74
|
Mathieu M, Odagiu L, Gaudot L, Daudelin JF, Melichar HJ, Lapointe R, Labrecque N. Inflammation enhances the vaccination potential of CD40-activated B cells in mice. Eur J Immunol 2016; 47:269-279. [PMID: 27873323 DOI: 10.1002/eji.201646568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/28/2016] [Accepted: 11/17/2016] [Indexed: 12/29/2022]
Abstract
Vaccination with antigen-pulsed CD40-activated B (CD40-B) cells can efficiently lead to the in vivo differentiation of naive CD8+ T cells into fully functional effectors. In contrast to bone marrow-derived dendritic cell (BMDC) vaccination, CD40-B cell priming does not allow for memory CD8+ T-cell generation but the reason for this deficiency is unknown. Here, we show that compared to BMDCs, murine CD40-B cells induce lower expression of several genes regulated by T-cell receptor signaling, costimulation, and inflammation (signals 1-3) in mouse T cells. The reduced provision of signals 1 and 2 by CD40-B cells can be explained by a reduction in the quality and duration of the interactions with naive CD8+ T cells as compared to BMDCs. Furthermore, CD40-B cells produce less inflammatory mediators, such as IL-12 and type I interferon, and increasing inflammation by coadministration of polyriboinosinic-polyribocytidylic acid with CD40-B-cell immunization allowed for the generation of long-lived and functional CD8+ memory T cells. In conclusion, it is possible to manipulate CD40-B-cell vaccination to promote the formation of long-lived functional CD8+ memory T cells, a key step before translating the use of CD40-B cells for therapeutic vaccination.
Collapse
Affiliation(s)
- Mélissa Mathieu
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, Québec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, University of Montreal, Montréal, Québec, Canada
| | - Livia Odagiu
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, Québec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, University of Montreal, Montréal, Québec, Canada
| | - Léa Gaudot
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, Québec, Canada
| | | | - Heather J Melichar
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, Québec, Canada.,Department of Medicine, University of Montreal, Montréal, Québec, Canada
| | - Réjean Lapointe
- Department of Medicine, University of Montreal, Montréal, Québec, Canada.,Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), University of Montreal and Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Nathalie Labrecque
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, Québec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, University of Montreal, Montréal, Québec, Canada.,Department of Medicine, University of Montreal, Montréal, Québec, Canada
| |
Collapse
|
75
|
Santos LC, Blair DA, Kumari S, Cammer M, Iskratsch T, Herbin O, Alexandropoulos K, Dustin ML, Sheetz MP. Actin polymerization-dependent activation of Cas-L promotes immunological synapse stability. Immunol Cell Biol 2016; 94:981-993. [PMID: 27359298 PMCID: PMC5121033 DOI: 10.1038/icb.2016.61] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 06/06/2016] [Accepted: 06/20/2016] [Indexed: 02/07/2023]
Abstract
The immunological synapse formed between a T-cell and an antigen-presenting cell is important for cell-cell communication during T-cell-mediated immune responses. Immunological synapse formation begins with stimulation of the T-cell receptor (TCR). TCR microclusters are assembled and transported to the center of the immunological synapse in an actin polymerization-dependent process. However, the physical link between TCR and actin remains elusive. Here we show that lymphocyte-specific Crk-associated substrate (Cas-L), a member of a force sensing protein family, is required for transport of TCR microclusters and for establishing synapse stability. We found that Cas-L is phosphorylated at TCR microclusters in an actin polymerization-dependent fashion. Furthermore, Cas-L participates in a positive feedback loop leading to amplification of Ca2+ signaling, inside-out integrin activation, and actomyosin contraction. We propose a new role for Cas-L in T-cell activation as a mechanical transducer linking TCR microclusters to the underlying actin network and coordinating multiple actin-dependent structures in the immunological synapse. Our studies highlight the importance of mechanotransduction processes in T-cell-mediated immune responses.
Collapse
Affiliation(s)
- Luís C Santos
- Department of Biological Sciences, Columbia UniversityNew YorkNYUSA
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
- Icahn Medical Institute, Mount Sinai School of MedicineNew YorkNYUSA
| | - David A Blair
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
| | - Sudha Kumari
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
| | - Michael Cammer
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
| | - Thomas Iskratsch
- Department of Biological Sciences, Columbia UniversityNew YorkNYUSA
| | - Olivier Herbin
- Icahn Medical Institute, Mount Sinai School of MedicineNew YorkNYUSA
| | | | - Michael L Dustin
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
- Kennedy Institute of Rheumatology, University of OxfordHeadingtonUK
| | - Michael P Sheetz
- Department of Biological Sciences, Columbia UniversityNew YorkNYUSA
| |
Collapse
|
76
|
Fiege JK, Beura LK, Burbach BJ, Shimizu Y. Adhesion- and Degranulation-Promoting Adapter Protein Promotes CD8 T Cell Differentiation and Resident Memory Formation and Function during an Acute Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:2079-89. [PMID: 27521337 PMCID: PMC5010998 DOI: 10.4049/jimmunol.1501805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 07/13/2016] [Indexed: 11/19/2022]
Abstract
During acute infections, naive Ag-specific CD8 T cells are activated and differentiate into effector T cells, most of which undergo contraction after pathogen clearance. A small population of CD8 T cells persists as memory to protect against future infections. We investigated the role of adhesion- and degranulation-promoting adapter protein (ADAP) in promoting CD8 T cell responses to a systemic infection. Naive Ag-specific CD8 T cells lacking ADAP exhibited a modest expansion defect early after Listeria monocytogenes or vesicular stomatitis virus infection but comparable cytolytic function at the peak of response. However, reduced numbers of ADAP-deficient CD8 T cells were present in the spleen after the peak of the response. ADAP deficiency resulted in a greater frequency of CD127(+) CD8 memory precursors in secondary lymphoid organs during the contraction phase. Reduced numbers of ADAP-deficient killer cell lectin-like receptor G1(-) CD8 resident memory T (TRM) cell precursors were present in a variety of nonlymphoid tissues at the peak of the immune response, and consequently the total numbers of ADAP-deficient TRM cells were reduced at memory time points. TRM cells that did form in the absence of ADAP were defective in effector molecule expression. ADAP-deficient TRM cells exhibited impaired effector function after Ag rechallenge, correlating with defects in their ability to form T cell-APC conjugates. However, ADAP-deficient TRM cells responded to TGF-β signals and recruited circulating memory CD8 T cells. Thus, ADAP regulates CD8 T cell differentiation events following acute pathogen challenge that are critical for the formation and selected functions of TRM cells in nonlymphoid tissues.
Collapse
Affiliation(s)
- Jessica K Fiege
- Department of Laboratory Medicine and Pathology, Center for Immunology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Lalit K Beura
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Brandon J Burbach
- Department of Laboratory Medicine and Pathology, Center for Immunology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Yoji Shimizu
- Department of Laboratory Medicine and Pathology, Center for Immunology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455; and
| |
Collapse
|
77
|
Gao X, Arpin C, Marvel J, Prokopiou SA, Gandrillon O, Crauste F. IL-2 sensitivity and exogenous IL-2 concentration gradient tune the productive contact duration of CD8(+) T cell-APC: a multiscale modeling study. BMC SYSTEMS BIOLOGY 2016; 10:77. [PMID: 27535120 PMCID: PMC4989479 DOI: 10.1186/s12918-016-0323-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/21/2016] [Indexed: 01/17/2023]
Abstract
Background The CD8+ T cell immune response fights acute infections by intracellular pathogens and, by generating an immune memory, enables immune responses against secondary infections. Activation of the CD8+ T cell immune response involves a succession of molecular events leading to modifications of CD8+ T cell population. To understand the endogenous and exogenous mechanisms controlling the activation of CD8+ T cells and to investigate the influence of early molecular events on the long-term cell population behavior, we developed a multiscale computational model. It integrates three levels of description: a Cellular Potts model describing the individual behavior of CD8+ T cells, a system of ordinary differential equations describing a decision-making molecular regulatory network at the intracellular level, and a partial differential equation describing the diffusion of IL-2 in the extracellular environment. Results We first calibrated the model parameters based on in vivo data and showed the model’s ability to reproduce early dynamics of CD8+ T cells in murine lymph nodes after influenza infection, both at the cell population and intracellular levels. We then showed the model’s ability to reproduce the proliferative responses of CD5hi and CD5lo CD8+ T cells to exogenous IL-2 under a weak TCR stimulation. This stressed the role of short-lasting molecular events and the relevance of explicitly describing both intracellular and cellular scale dynamics. Our results suggest that the productive contact duration of CD8+ T cell-APC is influenced by the sensitivity of individual CD8+ T cells to the activation signal and by the IL-2 concentration in the extracellular environment. Conclusions The multiscale nature of our model allows the reproduction and explanation of some acquired characteristics and functions of CD8+ T cells, and of their responses to multiple stimulation conditions, that would not be accessible in a classical description of cell population dynamics that would not consider intracellular dynamics. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0323-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuefeng Gao
- Inria team Dracula, Inria Antenne Lyon la Doua, Bâtiment CEI-2, 56 Boulevard Niels Bohr, 69603, Villeurbanne cedex, France
| | - Christophe Arpin
- Inserm, U1111, Lyon, F-69007, France.,CNRS, UMR5308, Lyon, F-69007, France.,Centre International de Recherche en Infectiologie, Université Lyon 1, Lyon, F-69007, France.,Ecole Normale Supérieure de Lyon, Lyon, F-69007, France
| | - Jacqueline Marvel
- Inserm, U1111, Lyon, F-69007, France.,CNRS, UMR5308, Lyon, F-69007, France.,Centre International de Recherche en Infectiologie, Université Lyon 1, Lyon, F-69007, France.,Ecole Normale Supérieure de Lyon, Lyon, F-69007, France
| | - Sotiris A Prokopiou
- Inria team Dracula, Inria Antenne Lyon la Doua, Bâtiment CEI-2, 56 Boulevard Niels Bohr, 69603, Villeurbanne cedex, France
| | - Olivier Gandrillon
- Inria team Dracula, Inria Antenne Lyon la Doua, Bâtiment CEI-2, 56 Boulevard Niels Bohr, 69603, Villeurbanne cedex, France. .,Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d'Italie Site Jacques Monod, F-69007, Lyon, France.
| | - Fabien Crauste
- Inria team Dracula, Inria Antenne Lyon la Doua, Bâtiment CEI-2, 56 Boulevard Niels Bohr, 69603, Villeurbanne cedex, France. .,Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5208, Institut Camille Jordan, 43 blvd. du 11 novembre 1918, F-69622, Villeurbanne cedex, France.
| |
Collapse
|
78
|
Baptista MAP, Keszei M, Oliveira M, Sunahara KKS, Andersson J, Dahlberg CIM, Worth AJ, Liedén A, Kuo IC, Wallin RPA, Snapper SB, Eidsmo L, Scheynius A, Karlsson MCI, Bouma G, Burns SO, Forsell MNE, Thrasher AJ, Nylén S, Westerberg LS. Deletion of Wiskott-Aldrich syndrome protein triggers Rac2 activity and increased cross-presentation by dendritic cells. Nat Commun 2016; 7:12175. [PMID: 27425374 PMCID: PMC4960314 DOI: 10.1038/ncomms12175] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 06/08/2016] [Indexed: 11/22/2022] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is caused by loss-of-function mutations in the WASp gene. Decreased cellular responses in WASp-deficient cells have been interpreted to mean that WASp directly regulates these responses in WASp-sufficient cells. Here, we identify an exception to this concept and show that WASp-deficient dendritic cells have increased activation of Rac2 that support cross-presentation to CD8(+) T cells. Using two different skin pathology models, WASp-deficient mice show an accumulation of dendritic cells in the skin and increased expansion of IFNγ-producing CD8(+) T cells in the draining lymph node and spleen. Specific deletion of WASp in dendritic cells leads to marked expansion of CD8(+) T cells at the expense of CD4(+) T cells. WASp-deficient dendritic cells induce increased cross-presentation to CD8(+) T cells by activating Rac2 that maintains a near neutral pH of phagosomes. Our data reveals an intricate balance between activation of WASp and Rac2 signalling pathways in dendritic cells.
Collapse
Affiliation(s)
- Marisa A. P. Baptista
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
- Institute for Virology and Immunobiology, University of Würzburg, 97078 Würzburg, Germany
| | - Marton Keszei
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Mariana Oliveira
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Karen K. S. Sunahara
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
- Experimental Physiopathology, Department of Sciences/Experimental Physiopatholgy, Medical School, University of São Paulo, São Paulo, Brazil
| | - John Andersson
- Department of Medicine Solna, Translational Immunology Unit, Karolinska Institutet and Karolinska University Hospital, Stockholm 171 76, Sweden
| | - Carin I. M. Dahlberg
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Austen J. Worth
- University College London Institute of Child Health, London WC1N 1EH, UK
| | - Agne Liedén
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden
| | - I-Chun Kuo
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, The National University Health System, Singapore 119228, Singapore
| | - Robert P. A. Wallin
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Scott B. Snapper
- Gastroenterology Division, Children's Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Liv Eidsmo
- Department of Medicine Solna, Dermatology and Venereology Unit, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Annika Scheynius
- Department of Medicine Solna, Translational Immunology Unit, Karolinska Institutet and Karolinska University Hospital, Stockholm 171 76, Sweden
| | - Mikael C. I. Karlsson
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Gerben Bouma
- University College London Institute of Child Health, London WC1N 1EH, UK
| | - Siobhan O. Burns
- University College London Institute of Child Health, London WC1N 1EH, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- University College London Institute of Immunity and Transplantation, London WC1E 6BT, UK
| | - Mattias N. E. Forsell
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
- Department of Clinical Microbiology, Division of Immunology, Umeå University, Umeå 901 87, Sweden
| | - Adrian J. Thrasher
- University College London Institute of Child Health, London WC1N 1EH, UK
| | - Susanne Nylén
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Lisa S. Westerberg
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
79
|
da Cunha A, Antoniazi Michelin M, Cândido Murta EF. Phenotypic profile of dendritic and T cells in the lymph node of Balb/C mice with breast cancer submitted to dendritic cells immunotherapy. Immunol Lett 2016; 177:25-37. [PMID: 27423825 DOI: 10.1016/j.imlet.2016.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/01/2016] [Accepted: 07/12/2016] [Indexed: 11/25/2022]
Abstract
Breast cancer (BC) is the most common malignant neoplasm and the cause of death by cancer among women worldwide. Its development influenced by various mutations that occur in the tumor cell and by the immune system's status, which has a direct influence on the tumor microenvironment and, consequently, on interactions with non-tumor cells involved in the immunological response. Strategies using dendritic cells (DCs) or antigen-presenting cells (APCs), therapeutic mode, in cancer have been developed for some time. The proper interaction between DCs and T cells upon antigen presentation is of greatest importance for an antitumor immune response activation. Thus, various receptors on the surface of T cells must be able to recognize ligands that are located on the surface of APCs. However, little is known about the real behavior and interaction forms of CDs and T cells after vaccination. Due to the crucial importance of DCs in an effective anti-tumor immune response activation and the search for compliant results in inducing this response by immunotherapies with DCs, the phenotypic profile of DCs and T cells in lymph nodes obtained from female Balb/C mice with breast cancer induced by 4T1 cells and DCs treated with vaccines was investigated. We evaluated through flow cytometry based on the surface and intracellular molecules marking; as well as the presence of cytokines and chemokines, IL-2, IL-4, IL-10, IL-12, IFN-γ, TNF-α and TGF-β in the supernatant of the culture of Balb/C lymph nodes by ELISA. The results show that the vaccination with DCs, in the maturation parameters used in this study, was able to stimulate the secretion of cytokines such as IFN-γ and IL-12 and inhibit the secretion of TGF-β and IL-10 in nodal lymph infiltrates, as well as co-stimulatory activating (CD86) and adhesion molecules in DCs and T cells LFA-1/ICAM-1 and inhibit the secretion of CTLA-4 present in lymph nodes. Facts that led to aTh1 profile polarization, immuno competent in relation to breast cancer. We indirectly evaluated the interaction between DCs and T cells dependent on the vaccination with DCs in tumor draining lymph nodes, in breast cancer in Balb/C mice and we believe that, this way, we will be able to achieve a model vaccine protocol in the future, based on the correct interaction between cells that enable the induction of anti-tumor effective response. Breast cancer (BC) is the most common malignant neoplasm and the cause of death by cancer among women worldwide. Its development influenced by various mutations that occur in the tumor cell and by the immune system's status, which has a direct influence on the tumor microenvironment and, consequently, on interactions with non-tumor cells involved in the immunological response. Strategies using dendritic cells (DCs) or antigen-presenting cells (APCs), therapeutic mode, in cancer have been developed for some time. The proper interaction between DCs and T cells upon antigen presentation is of greatest importance for an antitumor immune response activation. Thus, various receptors on the surface of T cells must be able to recognize ligands that are located on the surface of APCs. However, little is known about the real behavior and interaction forms of DCs and T cells after vaccination. Due to the crucial importance of DCs in an effective anti-tumor immune response activation and the search for compliant results in inducing this response by immunotherapies with DCs, the phenotypic profile of DCs and T cells in lymph nodes obtained from female Balb/C mice with breast cancer induced by 4T1 cells and DCs treated with vaccines was investigated. We evaluated through flow cytometry based on the surface and intracellular molecules marking; as well as the presence of cytokines and chemokines, IL-2, IL-4, IL-10, IL-12, IFN-γ, TNF-α and TGF-β in the supernatant of the culture of Balb/C lymph nodes by ELISA. The results show that the vaccination with DCs, in the maturation parameters used in this study, was able to stimulate the secretion of cytokines such as IFN-γ and IL-12 and inhibit the secretion of TGF-β and IL-10 in nodal lymph infiltrates, as well as co-stimulatory activating (CD86) and adhesion molecules in DCs and T cells LFA-1/ICAM-1 and inhibit the secretion of CTLA-4 present in lymph nodes. Facts that led to aTh1 profile polarization, immuno competent in relation to breast cancer. We indirectly evaluated the interaction between DCs and T cells dependent on the vaccination with DCs in tumor draining lymph nodes, in breast cancer in Balb/C mice and we believe that, this way, we will be able to achieve a model vaccine protocol in the future, based on the correct interaction between cells that enable the induction of anti-tumor effective response.
Collapse
Affiliation(s)
- Alessandra da Cunha
- Research Oncology Institute (IPON), Federal University of the Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-440, Brazil
| | - Marcia Antoniazi Michelin
- Discipline of Immunology, Federal University of the Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-440, Brazil
| | - Eddie Fernando Cândido Murta
- Discipline of Gynecology and Obstetrics, Federal University of the Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-440, Brazil.
| |
Collapse
|
80
|
Capece T, Kim M. The Role of Lymphatic Niches in T Cell Differentiation. Mol Cells 2016; 39:515-23. [PMID: 27306645 PMCID: PMC4959015 DOI: 10.14348/molcells.2016.0089] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/21/2016] [Accepted: 05/24/2016] [Indexed: 11/27/2022] Open
Abstract
Long-term immunity to many viral and bacterial pathogens requires CD8(+) memory T cell development, and the induction of long-lasting CD8(+) memory T cells from a naïve, undifferentiated state is a major goal of vaccine design. Formation of the memory CD8(+) T cell compartment is highly dependent on the early activation cues received by naïve CD8(+) T cells during primary infection. This review aims to highlight the cellularity of various niches within the lymph node and emphasize recent evidence suggesting that distinct types of T cell activation and differentiation occur within different immune contexts in lymphoid organs.
Collapse
Affiliation(s)
- Tara Capece
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642,
USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642,
USA
| |
Collapse
|
81
|
Emerging therapies provide new opportunities to reshape the multifaceted interactions between the immune system and lymphoma cells. Leukemia 2016; 30:1805-15. [PMID: 27389058 DOI: 10.1038/leu.2016.161] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 12/21/2022]
Abstract
The acquisition of a complete neoplastic phenotype requires cancer cells to develop escape mechanisms from the host immune system. This phenomenon, commonly referred to as 'immune evasion,' represents a hallmark of cancers and results from a Darwinian selection of the fittest tumor clones. First reported in solid tumors, cancer immunoescape characterizes several hematological malignancies. The biological bases of cancer immunoescape have recently been disclosed and include: (i) impaired human leukocyte antigen-mediated cancer cell recognition (B2M, CD58, CTIIA, CD80/CD86, CD28 and CTLA-4 mutations); (ii) deranged apoptotic mechanisms (reduced pro-apoptotic signals and/or increased expression of anti-apoptotic molecules); and (iii) changes in the tumor microenvironment involving regulatory T cells and tumor-associated macrophages. These immune-escape mechanisms characterize both Hodgkin and non-Hodgkin (B and T cell) lymphomas and represent a promising target for new anti-tumor therapies. In the present review, the principles of cancer immunoescape and their role in human lymphomagenesis are illustrated. Current therapies targeting these pathways and possible applications for lymphoma treatment are also addressed.
Collapse
|
82
|
Palmer E, Drobek A, Stepanek O. Opposing effects of actin signaling and LFA-1 on establishing the affinity threshold for inducing effector T-cell responses in mice. Eur J Immunol 2016; 46:1887-901. [PMID: 27188212 DOI: 10.1002/eji.201545909] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 05/02/2016] [Accepted: 05/13/2016] [Indexed: 11/10/2022]
Abstract
Mature CD8(+) T cells use a narrow antigen affinity threshold to generate tissue-infiltrating cytotoxic effector T cells and induce autoimmune pathology, but the mechanisms that establish this antigen affinity threshold are poorly understood. Only antigens with affinities above the threshold induce stable contacts with APCs, polarization of a T cell, and asymmetric T-cell division. Previously published data indicate that LFA-1 inside-out signaling might be involved in establishing the antigen affinity threshold. Here, we show that subthreshold antigens weakly activate all major distal TCR signaling pathways. Low-affinity antigens are more dependent on LFA-1 than suprathreshold antigens. Moreover, augmenting the inside-out signaling by hyperactive Rap1 does not increase responses to the subthreshold antigens. Thus, LFA-1 signaling does not contribute to the affinity-based antigen discrimination. However, we found that subthreshold antigens do not induce actin rearrangement toward an APC, mediated by Rho-family GTPases, Cdc42, and Rac. Our data suggest that Rac and Cdc42 contribute to the establishment of the antigen affinity threshold in CD8(+) T cells by enhancing responses to high-affinity antigens, or by reducing the responses to low-affinity antigens.
Collapse
Affiliation(s)
- Ed Palmer
- Departments of Biomedicine and Nephrology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Ales Drobek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Stepanek
- Departments of Biomedicine and Nephrology, University Hospital Basel and University of Basel, Basel, Switzerland.,Laboratory of Adaptive Immunity, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
83
|
Katakai T, Kinashi T. Microenvironmental Control of High-Speed Interstitial T Cell Migration in the Lymph Node. Front Immunol 2016; 7:194. [PMID: 27242799 PMCID: PMC4865483 DOI: 10.3389/fimmu.2016.00194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/02/2016] [Indexed: 12/30/2022] Open
Abstract
T cells are highly concentrated in the lymph node (LN) paracortex, which serves an important role in triggering adoptive immune responses. Live imaging using two-photon laser scanning microscopy revealed vigorous and non-directional T cell migration within this area at average velocity of more than 10 μm/min. Active interstitial T cell movement is considered to be crucial for scanning large numbers of dendritic cells (DCs) to find rare cognate antigens. However, the mechanism by which T cells achieve such high-speed movement in a densely packed, dynamic tissue environment is not fully understood. Several new findings suggest that fibroblastic reticular cells (FRCs) and DCs control T cell movement in a multilateral manner. Chemokines and lysophosphatidic acid produced by FRCs cooperatively promote the migration, while DCs facilitate LFA-1-dependent motility via expression of ICAM-1. Furthermore, the highly dense and confined microenvironment likely plays a key role in anchorage-independent motility. We propose that T cells dynamically switch between two motility modes; anchorage-dependent and -independent manners. Unique tissue microenvironment and characteristic migration modality of T cells cooperatively generate high-speed interstitial movement in the LN.
Collapse
Affiliation(s)
- Tomoya Katakai
- Department of Immunology, Graduate School of Medical and Dental Sciences, Niigata University , Niigata , Japan
| | - Tatsuo Kinashi
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University , Hirakata , Japan
| |
Collapse
|
84
|
Moreau HD, Bogle G, Bousso P. A virtual lymph node model to dissect the requirements for T-cell activation by synapses and kinapses. Immunol Cell Biol 2016; 94:680-8. [PMID: 27089942 PMCID: PMC4980574 DOI: 10.1038/icb.2016.36] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 12/18/2022]
Abstract
The initiation of T-cell responses in lymph nodes requires T cells to integrate signals delivered by dendritic cells (DCs) during long-lasting contacts (synapses) or more transient interactions (kinapses). However, it remains extremely challenging to understand how a specific sequence of contacts established by T cells ultimately dictates T-cell fate. Here, we have coupled a computational model of T-cell migration and interactions with DCs with a real-time, flow cytometry-like representation of T-cell activation. In this model, low-affinity peptides trigger T-cell proliferation through kinapses but we show that this process is only effective under conditions of high DC densities and prolonged antigen availability. By contrast, high-affinity peptides favor synapse formation and a vigorous proliferation under a wide range of antigen presentation conditions. In line with the predictions, decreasing the DC density in vivo selectively abolished proliferation induced by the low-affinity peptide. Finally, our results suggest that T cells possess a biochemical memory of previous stimulations of at least 1–2 days. We propose that the stability of T-cell–DC interactions, apart from their signaling potency, profoundly influences the robustness of T-cell activation. By offering the ability to control parameters that are difficult to manipulate experimentally, the virtual lymph node model provides new possibilities to tackle the fundamental mechanisms that regulate T-cell responses elicited by infections or vaccines.
Collapse
Affiliation(s)
- Hélène D Moreau
- Institut Pasteur, Dynamics of Immune Responses Unit, Paris, France.,INSERM U1223, Paris, France
| | - Gib Bogle
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Philippe Bousso
- Institut Pasteur, Dynamics of Immune Responses Unit, Paris, France.,INSERM U1223, Paris, France
| |
Collapse
|
85
|
Askew D, Su CA, Barkauskas DS, Dorand RD, Myers J, Liou R, Nthale J, Huang AY. Transient Surface CCR5 Expression by Naive CD8+ T Cells within Inflamed Lymph Nodes Is Dependent on High Endothelial Venule Interaction and Augments Th Cell-Dependent Memory Response. THE JOURNAL OF IMMUNOLOGY 2016; 196:3653-64. [PMID: 26994221 DOI: 10.4049/jimmunol.1501176] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 02/24/2016] [Indexed: 11/19/2022]
Abstract
In inflamed lymph nodes, Ag-specific CD4(+) and CD8(+) T cells encounter Ag-bearing dendritic cells and, together, this complex enhances the release of CCL3 and CCL4, which facilitate additional interaction with naive CD8(+) T cells. Although blocking CCL3 and CCL4 has no effect on primary CD8(+) T cell responses, it dramatically impairs the development of memory CD8(+) T cells upon Ag rechallenge. Despite the absence of detectable surface CCR5 expression on circulating native CD8(+) T cells, these data imply that naive CD8(+) T cells are capable of expressing surface CCR5 prior to cognate Ag-induced TCR signaling in inflamed lymph nodes; however, the molecular mechanisms have not been characterized to date. In this study, we show that CCR5, the receptor for CCL3 and CCL4, can be transiently upregulated on a subset of naive CD8(+) T cells and that this upregulation is dependent on direct contact with the high endothelial venule in inflamed lymph node. Binding of CD62L and CD11a on T cells to their ligands CD34 and CD54 on the high endothelial venule can be enhanced during inflammation. This enhanced binding and subsequent signaling promote the translocation of CCR5 molecules from intracellular vesicles to the surface of the CD8(+) T cell. The upregulation of CCR5 on the surface of the CD8(+) T cells increases the number of contacts with Ag-bearing dendritic cells, which ultimately results in increased CD8(+) T cell response to Ag rechallenge.
Collapse
Affiliation(s)
- David Askew
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106;
| | - Charles A Su
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland OH 44195; and
| | - Deborah S Barkauskas
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - R Dixon Dorand
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Jay Myers
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Rachel Liou
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Joseph Nthale
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Alex Y Huang
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
86
|
Spranger S, Sivan A, Corrales L, Gajewski TF. Tumor and Host Factors Controlling Antitumor Immunity and Efficacy of Cancer Immunotherapy. Adv Immunol 2016; 130:75-93. [PMID: 26923000 DOI: 10.1016/bs.ai.2015.12.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite recent clinical advances in immunotherapy, a fraction of cancer patients fails to respond to these interventions. Evidence from preclinical mouse models as well as clinical samples has provided evidence that the extent of activated T cell infiltration within the tumor microenvironment is associated with clinical response to immunotherapies including checkpoint blockade. Therefore, understanding the molecular mechanisms mediating the lack of T cell infiltration into the tumor microenvironment will be instrumental for the development of new therapeutic strategies to render those patients immunotherapy responsive. Recent data have suggested that major sources of intersubject heterogeneity include differences in somatic mutations in specific oncogene pathways between cancers of individual subjects and also environmental factors including commensal microbial composition. Successful identification of such causal factors should lead to new therapeutic approaches that may facilitate T cell entry into noninflamed tumors and expand the fraction of patients capable of responding to novel immunotherapies.
Collapse
Affiliation(s)
| | - Ayelet Sivan
- The University of Chicago, Chicago, IL, United States
| | | | | |
Collapse
|
87
|
PI3Kδ promotes CD4(+) T-cell interactions with antigen-presenting cells by increasing LFA-1 binding to ICAM-1. Immunol Cell Biol 2016; 94:486-95. [PMID: 26740009 PMCID: PMC4829101 DOI: 10.1038/icb.2016.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/30/2015] [Accepted: 12/30/2015] [Indexed: 12/15/2022]
Abstract
Activation of T lymphocytes by peptide/major histocompatibility complex on antigen-presenting cells (APCs) involves dynamic contacts between the two cells, during which T cells undergo marked morphological changes. These interactions are facilitated by integrins. Activation of the T cells increases the binding of the integrin lymphocyte function-associated antigen 1 (LFA-1) expressed by T cells to intercellular adhesion molecule (ICAM)-1 and ICAM-2 expressed by APCs. The signalling pathways that control integrin affinities are incompletely defined. The phosphoinositide 3-kinases (PI3Ks) generate second-messenger signalling molecules that control cell growth, proliferation, differentiation and trafficking. Here we show that in T cells, PI3Kδ attenuates the activation of Rac1, but sustains the activation of Rap1. Consequently, PI3Kδ increases LFA-1-dependent adhesion to form stable conjugates with APCs. Increased Rap1 activity and LFA-1 adhesion were only in part mediated by the downstream kinase Akt, suggesting the involvement of additional phosphatidylinositol(3,4,5)P3-binding proteins. These results establish a link between PI3K activity, cytoskeletal changes and integrin binding and help explain the impaired T-cell-dependent immune responses in PI3Kδ-deficient mice.
Collapse
|
88
|
Huranova M, Stepanek O. Role of actin cytoskeleton at multiple levels of T cell activation. AIMS MOLECULAR SCIENCE 2016. [DOI: 10.3934/molsci.2016.4.585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
89
|
Gottrand G, Courau T, Thomas‐Vaslin V, Prevel N, Vazquez T, Ruocco MG, Lambrecht B, Bellier B, Colombo BM, Klatzmann D. Regulatory T-cell development and function are impaired in mice lacking membrane expression of full length intercellular adhesion molecule-1. Immunology 2015; 146:657-70. [PMID: 26370005 PMCID: PMC4693902 DOI: 10.1111/imm.12533] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 08/17/2015] [Accepted: 09/10/2015] [Indexed: 12/15/2022] Open
Abstract
To further investigate the contribution of intercellular adhesion molecule-1 (ICAM-1) to adaptive immune responses, we analysed T-cell development and function in mice lacking full-length ICAM-1 (ICAM-1(tm1Jcgr) ). Compared with wild-type (ICAM-1(WT) ) mice, ICAM-1(tm1Jcgr) mice have impaired thymocyte development. Proportions and numbers of double negative, double positive, mature CD4(+) and CD8(+) thymocytes, as well as of regulatory T (Treg) cells were also significantly decreased. In the periphery, ICAM-1(tm1Jcgr) mice had significantly decreased proportions and numbers of naive and activated/memory CD4(+) and CD8(+) T cells, as well as of Treg cells, in lymph nodes but not in the spleen. In vitro activation of CD4(+) and CD8(+) T cells from ICAM-1(tm1Jcgr) mice with anti-CD3 antibodies and antigen-presenting cells (APCs) resulted in a significantly weaker proliferation, whereas proliferation induced with anti-CD3 and anti-CD28 antibody-coated beads was normal. In vivo immunization of ICAM-1(tm1Jcgr) mice resulted in normal generation of specific effector and memory immune responses that protect against a viral challenge. However, contrary to ICAM-1(WT) mice, immunization-induced specific effectors could not eradicate immunogen-expressing tumours. Treg cells from ICAM-1(tm1Jcgr) mice have abnormal activation and proliferation induced by anti-CD3 antibody and APCs, and have markedly decreased suppressive activity in vitro. In contrast to ICAM-1(WT) mice, they were unable to control experimentally induced colitis in vivo. Hence, our results further highlight the pleiotropic role of ICAM-1 in T-cell-dependent immune responses, with a major role in Treg cell development and suppressive function.
Collapse
Affiliation(s)
- Gaëlle Gottrand
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
| | - Tristan Courau
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
| | - Véronique Thomas‐Vaslin
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
| | - Nicolas Prevel
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
| | - Thomas Vazquez
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
| | - Maria Grazia Ruocco
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
| | | | - Bertrand Bellier
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
| | - Bruno M. Colombo
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
- Institute of Systems and Synthetic Biology (iSSB)CNRS FRE3561Université d'Evry‐val d'EssonneGenopole91058EvryFrance
| | - David Klatzmann
- Immunology‐Immunopathology‐ImmunotherapyUPMC Univ Paris 06UMRS_959Sorbonne UniversitésParisFrance
- Immunology‐Immunopathology‐ImmunotherapyFRE3632CNRSParisFrance
- Immunology‐Immunopathology‐ImmunotherapyUMRS_959INSERMParisFrance
| |
Collapse
|
90
|
Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre ML, Chang EB, Gajewski TF. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015; 350:1084-9. [PMID: 26541606 DOI: 10.1126/science.aac4255] [Citation(s) in RCA: 2618] [Impact Index Per Article: 261.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 10/09/2015] [Indexed: 12/11/2022]
Abstract
T cell infiltration of solid tumors is associated with favorable patient outcomes, yet the mechanisms underlying variable immune responses between individuals are not well understood. One possible modulator could be the intestinal microbiota. We compared melanoma growth in mice harboring distinct commensal microbiota and observed differences in spontaneous antitumor immunity, which were eliminated upon cohousing or after fecal transfer. Sequencing of the 16S ribosomal RNA identified Bifidobacterium as associated with the antitumor effects. Oral administration of Bifidobacterium alone improved tumor control to the same degree as programmed cell death protein 1 ligand 1 (PD-L1)-specific antibody therapy (checkpoint blockade), and combination treatment nearly abolished tumor outgrowth. Augmented dendritic cell function leading to enhanced CD8(+) T cell priming and accumulation in the tumor microenvironment mediated the effect. Our data suggest that manipulating the microbiota may modulate cancer immunotherapy.
Collapse
Affiliation(s)
- Ayelet Sivan
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Leticia Corrales
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Nathaniel Hubert
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jason B Williams
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | | | - Zachary M Earley
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Franco W Benyamin
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Yuk Man Lei
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | - Eugene B Chang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Thomas F Gajewski
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA. Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
91
|
Palmer DC, Guittard GC, Franco Z, Crompton JG, Eil RL, Patel SJ, Ji Y, Van Panhuys N, Klebanoff CA, Sukumar M, Clever D, Chichura A, Roychoudhuri R, Varma R, Wang E, Gattinoni L, Marincola FM, Balagopalan L, Samelson LE, Restifo NP. Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance. J Exp Med 2015; 212:2095-113. [PMID: 26527801 PMCID: PMC4647263 DOI: 10.1084/jem.20150304] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 09/11/2015] [Indexed: 01/17/2023] Open
Abstract
Palmer et al. find that Cish, a member of the SOCS family, is induced by TCR stimulation in CD8+ T cells and inhibits their functional avidity against tumor. The authors uncover a novel mechanism of suppression for a SOCS member. Improving the functional avidity of effector T cells is critical in overcoming inhibitory factors within the tumor microenvironment and eliciting tumor regression. We have found that Cish, a member of the suppressor of cytokine signaling (SOCS) family, is induced by TCR stimulation in CD8+ T cells and inhibits their functional avidity against tumors. Genetic deletion of Cish in CD8+ T cells enhances their expansion, functional avidity, and cytokine polyfunctionality, resulting in pronounced and durable regression of established tumors. Although Cish is commonly thought to block STAT5 activation, we found that the primary molecular basis of Cish suppression is through inhibition of TCR signaling. Cish physically interacts with the TCR intermediate PLC-γ1, targeting it for proteasomal degradation after TCR stimulation. These findings establish a novel targetable interaction that regulates the functional avidity of tumor-specific CD8+ T cells and can be manipulated to improve adoptive cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yun Ji
- National Cancer Institute, Bethesda, MD 20892
| | | | | | | | - David Clever
- National Cancer Institute, Bethesda, MD 20892 Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, OH 43210
| | | | | | - Rajat Varma
- National Institute of Allergy and Infectious Disease, Bethesda, MD 20892
| | - Ena Wang
- Sidra Medical and Research Center, Doha, Qatar
| | | | | | | | | | | |
Collapse
|
92
|
Stein JV. T Cell Motility as Modulator of Interactions with Dendritic Cells. Front Immunol 2015; 6:559. [PMID: 26579132 PMCID: PMC4629691 DOI: 10.3389/fimmu.2015.00559] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/19/2015] [Indexed: 01/13/2023] Open
Abstract
It is well established that the balance of costimulatory and inhibitory signals during interactions with dendritic cells (DCs) determines T cell transition from a naïve to an activated or tolerant/anergic status. Although many of these molecular interactions are well reproduced in reductionist in vitro assays, the highly dynamic motility of naïve T cells in lymphoid tissue acts as an additional lever to fine-tune their activation threshold. T cell detachment from DCs providing suboptimal stimulation allows them to search for DCs with higher levels of stimulatory signals, while storing a transient memory of short encounters. In turn, adhesion of weakly reactive T cells to DCs presenting peptides presented on major histocompatibility complex with low affinity is prevented by lipid mediators. Finally, controlled recruitment of CD8(+) T cells to cognate DC-CD4(+) T cell clusters shapes memory T cell formation and the quality of the immune response. Dynamic physiological lymphocyte motility therefore constitutes a mechanism to mitigate low avidity T cell activation and to improve the search for "optimal" DCs, while contributing to peripheral tolerance induction in the absence of inflammation.
Collapse
Affiliation(s)
- Jens V Stein
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| |
Collapse
|
93
|
Ramsingh AI, Manley K, Rong Y, Reilly A, Messer A. Transcriptional dysregulation of inflammatory/immune pathways after active vaccination against Huntington's disease. Hum Mol Genet 2015; 24:6186-97. [PMID: 26307082 PMCID: PMC4599676 DOI: 10.1093/hmg/ddv335] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/04/2015] [Accepted: 08/10/2015] [Indexed: 01/01/2023] Open
Abstract
Immunotherapy, both active and passive, is increasingly recognized as a powerful approach to a wide range of diseases, including Alzheimer's and Parkinson's. Huntington's disease (HD), an autosomal dominant disorder triggered by misfolding of huntingtin (HTT) protein with an expanded polyglutamine tract, could also benefit from this approach. Individuals can be identified genetically at the earliest stages of disease, and there may be particular benefits to a therapy that can target peripheral tissues in addition to brain. In this active vaccination study, we first examined safety and immunogenicity for a broad series of peptide, protein and DNA plasmid immunization protocols, using fragment (R6/1), and knock-in (zQ175) models. No safety issues were found. The strongest and most uniform immune response was to a combination of three non-overlapping HTT Exon1 coded peptides, conjugated to KLH, delivered with alum adjuvant. An N586-82Q plasmid, delivered via gene gun, also showed ELISA responses, mainly in the zQ175 strain, but with more variability, and less robust responses in HD compared with wild-type controls. Transcriptome profiling of spleens from the triple peptide-immunized cohort showed substantial HD-specific differences including differential activation of genes associated with innate immune responses, absence of negative feedback control of gene expression by regulators, a temporal dysregulation of innate immune responses and transcriptional repression of genes associated with memory T cell responses. These studies highlight critical issues for immunotherapy and HD disease management in general.
Collapse
Affiliation(s)
- Arlene I Ramsingh
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and
| | - Kevin Manley
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and
| | - Yinghui Rong
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and
| | - Andrew Reilly
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and
| | - Anne Messer
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY, USA
| |
Collapse
|
94
|
Ng D, Maître B, Cummings D, Lin A, Ward LA, Rahbar R, Mossman KL, Ohashi PS, Gommerman JL. A Lymphotoxin/Type I IFN Axis Programs CD8+T Cells To Infiltrate a Self-Tissue and Propagate Immunopathology. THE JOURNAL OF IMMUNOLOGY 2015; 195:4650-9. [DOI: 10.4049/jimmunol.1501053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/18/2015] [Indexed: 01/05/2023]
|
95
|
Tabdanov E, Gondarenko S, Kumari S, Liapis A, Dustin ML, Sheetz MP, Kam LC, Iskratsch T. Micropatterning of TCR and LFA-1 ligands reveals complementary effects on cytoskeleton mechanics in T cells. Integr Biol (Camb) 2015; 7:1272-84. [PMID: 26156536 PMCID: PMC4593733 DOI: 10.1039/c5ib00032g] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The formation of the immunological synapse between a T cell and the antigen-presenting cell (APC) is critically dependent on actin dynamics, downstream of T cell receptor (TCR) and integrin (LFA-1) signalling. There is also accumulating evidence that mechanical forces, generated by actin polymerization and/or myosin contractility regulate T cell signalling. Because both receptor pathways are intertwined, their contributions towards the cytoskeletal organization remain elusive. Here, we identify the specific roles of TCR and LFA-1 by using a combination of micropatterning to spatially separate signalling systems and nanopillar arrays for high-precision analysis of cellular forces. We identify that Arp2/3 acts downstream of TCRs to nucleate dense actin foci but propagation of the network requires LFA-1 and the formin FHOD1. LFA-1 adhesion enhances actomyosin forces, which in turn modulate actin assembly downstream of the TCR. Together our data shows a mechanically cooperative system through which ligands presented by an APC modulate T cell activation.
Collapse
Affiliation(s)
- Erdem Tabdanov
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Sasha Gondarenko
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Sudha Kumari
- Department of Pathology, New York University, New York, NY, USA
| | | | - Michael L. Dustin
- Department of Pathology, New York University, New York, NY, USA
- Kennedy Institute of Rheumatology, University of Oxford, Headington, Oxon, UK
| | - Michael P. Sheetz
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Lance C. Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Thomas Iskratsch
- Department of Biological Sciences, Columbia University, New York, NY, USA
| |
Collapse
|
96
|
Kallikourdis M, Viola A, Benvenuti F. Human Immunodeficiencies Related to Defective APC/T Cell Interaction. Front Immunol 2015; 6:433. [PMID: 26379669 PMCID: PMC4551858 DOI: 10.3389/fimmu.2015.00433] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 08/09/2015] [Indexed: 11/13/2022] Open
Abstract
The primary event for initiating adaptive immune responses is the encounter between T lymphocytes and antigen presenting cells (APCs) in the T cell area of secondary lymphoid organs and the formation of highly organized intercellular junctions referred to as immune synapses (IS). In vivo live-cell imaging of APC-T cell interactions combined to functional studies unveiled that T cell fate is dictated, in large part, by the stability of the initial contact. Immune cell interaction is equally important during delivery of T cell help to B cells and for the killing of target cells by cytotoxic T cells and NK cells. The critical role of contact dynamics and synapse stability on the immune response is well illustrated by human immune deficiencies in which disease pathogenesis is linked to altered adhesion or defective cross-talk between the synaptic partners. The Wiskott-Aldrich syndrome (WAS) is a severe primary immunodeficiency caused by mutations in the Wiskott-Aldrich syndrome protein (WASp), a scaffold that promotes actin polymerization and links TCR stimulation to T cell activation. Absence or mutations in WASp affects intercellular APC-T cell communications by interfering with multiple mechanisms on both sides of the IS. The warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome is caused by mutations in CXCR4, a chemokine receptor that in mutant form leads to impairment of APC-T cell interactions. Present evidences suggest that other recently characterized primary immune deficiencies caused by mutation in genes linked to actin cytoskeletal reorganization, such as WIP and DOCK8, may also depend on altered synapse stability. Here, we will discuss in details the mechanisms of disturbed APC-T cell interactions in WAS and WHIM. Moreover, we will summarize the evidence pointing to a compromised conjugate formation in WIP, DOCK8, and X-linked lymphoproliferative syndrome.
Collapse
Affiliation(s)
- Marinos Kallikourdis
- Humanitas University , Rozzano , Italy ; Adaptive Immunity Laboratory, Humanitas Clinical and Research Center , Rozzano , Italy
| | | | - Federica Benvenuti
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology , Trieste , Italy
| |
Collapse
|
97
|
Nassef Kadry Naguib Roufaiel M, Wells JW, Steptoe RJ. Impaired T-Cell Function in B-Cell Lymphoma: A Direct Consequence of Events at the Immunological Synapse? Front Immunol 2015; 6:258. [PMID: 26082776 PMCID: PMC4451642 DOI: 10.3389/fimmu.2015.00258] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/11/2015] [Indexed: 12/21/2022] Open
Abstract
Tumors can escape immune destruction through the development of antigen loss variants and loss of antigen processing/presentation pathways, thereby rendering them invisible to T cells. Alternatively, mechanisms of peripheral T-cell tolerance that would normally be important for protection from the development of autoimmunity may also be co-opted to (i) generate an immuno-inhibitory tumor environment, (ii) promote development of regulatory cell populations, or (iii) cell-intrinsically inactivate tumor-specific T cells. Emerging evidence suggests that T-cell function is impaired in hematological malignancies, which may manifest from cognate interactions between T cells and the tumor. The immunological synapse forms the cognate T-cell and antigen-presenting cell interaction and is the site where key signalling events, including those delivered by co-inhibitory receptors, that determine the fate of T cells occur. Here, we review evidence that events at the immune synapse between T cells and malignant B cells and alterations in immune synapse function may contribute to loss of T-cell function in B-cell malignancies.
Collapse
Affiliation(s)
- Marian Nassef Kadry Naguib Roufaiel
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute , Brisbane, QLD , Australia
| | - James W Wells
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute , Brisbane, QLD , Australia
| | - Raymond J Steptoe
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute , Brisbane, QLD , Australia
| |
Collapse
|
98
|
A dominant role for the methyl-CpG-binding protein Mbd2 in controlling Th2 induction by dendritic cells. Nat Commun 2015; 6:6920. [PMID: 25908537 PMCID: PMC4413429 DOI: 10.1038/ncomms7920] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/13/2015] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) direct CD4(+) T-cell differentiation into diverse helper (Th) subsets that are required for protection against varied infections. However, the mechanisms used by DCs to promote Th2 responses, which are important both for immunity to helminth infection and in allergic disease, are currently poorly understood. We demonstrate a key role for the protein methyl-CpG-binding domain-2 (Mbd2), which links DNA methylation to repressive chromatin structure, in regulating expression of a range of genes that are associated with optimal DC activation and function. In the absence of Mbd2, DCs display reduced phenotypic activation and a markedly impaired capacity to initiate Th2 immunity against helminths or allergens. These data identify an epigenetic mechanism that is central to the activation of CD4(+) T-cell responses by DCs, particularly in Th2 settings, and reveal methyl-CpG-binding proteins and the genes under their control as possible therapeutic targets for type-2 inflammation.
Collapse
|
99
|
Comrie WA, Li S, Boyle S, Burkhardt JK. The dendritic cell cytoskeleton promotes T cell adhesion and activation by constraining ICAM-1 mobility. ACTA ACUST UNITED AC 2015; 208:457-73. [PMID: 25666808 PMCID: PMC4332244 DOI: 10.1083/jcb.201406120] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Integrity of the dendritic cell (DC) actin cytoskeleton is essential for T cell priming, but the underlying mechanisms are poorly understood. We show that the DC F-actin network regulates the lateral mobility of intracellular cell adhesion molecule 1 (ICAM-1), but not MHCII. ICAM-1 mobility and clustering are regulated by maturation-induced changes in the expression and activation of moesin and α-actinin-1, which associate with actin filaments and the ICAM-1 cytoplasmic domain. Constrained ICAM-1 mobility is important for DC function, as DCs expressing a high-mobility ICAM-1 mutant lacking the cytoplasmic domain exhibit diminished antigen-dependent conjugate formation and T cell priming. These defects are associated with inefficient induction of leukocyte functional antigen 1 (LFA-1) affinity maturation, which is consistent with a model in which constrained ICAM-1 mobility opposes forces on LFA-1 exerted by the T cell cytoskeleton, whereas ICAM-1 clustering enhances valency and further promotes ligand-dependent LFA-1 activation. Our results reveal an important new mechanism through which the DC cytoskeleton regulates receptor activation at the immunological synapse.
Collapse
Affiliation(s)
- William A Comrie
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19102
| | - Shuixing Li
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19102
| | - Sarah Boyle
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19102
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19102
| |
Collapse
|
100
|
Mitsui A, Tada Y, Shibata S, Kamata M, Hau C, Asahina A, Sato S. Deficiency of both L-selectin and ICAM-1 exacerbates imiquimod-induced psoriasis-like skin inflammation through increased infiltration of antigen presenting cells. Clin Immunol 2015; 157:43-55. [PMID: 25572533 DOI: 10.1016/j.clim.2014.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/06/2014] [Accepted: 12/19/2014] [Indexed: 12/30/2022]
Abstract
To assess the role of inter-cellular adhesion molecule (ICAM)-1 and L-selectin in psoriasis pathogenic process, we examined the psoriasiform skin inflammation triggered by imiquimod, a toll-like receptor 7/8 agonist, in mice lacking ICAM-1 (ICAM-1(-/-)), L-selectin (L-selectin(-/-)), or both (L-selectin/ICAM-1(-/-)). Disease severity was significantly reduced in ICAM-1(-/-) and L-selectin(-/-) mice compared with wild type mice, while it was exacerbated in L-selectin/ICAM-1(-/-) mice. Cutaneous interleukin (IL)-17A, IL-23, and tumor necrosis factor (TNF)-α expression was increased in L-selectin/ICAM-1(-/-) mice compared with wild type mice. Furthermore, only L-selectin/ICAM-1(-/-) mice was refractory to anti-TNF-α antibody treatment. The skin lesion from L-selectin/ICAM-1(-/-) mice showed augmented E-selectin expression compared with ICAM-1(-/-) and L-selectin(-/-) mice, and augmented E-selectin ligand-1 expression compared with wild type mice. The current study demonstrates that although ICAM-1 and L-selectin regulate psoriasiform inflammation, deleting both L-selectin and ICAM-1 simultaneously would rather induce refractory skin inflammation, due to compensatory up-regulation of other adhesion molecules.
Collapse
Affiliation(s)
- Aya Mitsui
- Department of Dermatology, Faculty of Medicine, University of Tokyo, Japan
| | - Yayoi Tada
- Department of Dermatology, Faculty of Medicine, University of Tokyo, Japan.
| | - Sayaka Shibata
- Department of Dermatology, Faculty of Medicine, University of Tokyo, Japan
| | - Masahiro Kamata
- Department of Dermatology, Faculty of Medicine, University of Tokyo, Japan
| | - Carren Hau
- Department of Dermatology, Faculty of Medicine, University of Tokyo, Japan
| | - Akihiko Asahina
- Department of Dermatology, Sagamihara National Hospital, Kanagawa, Japan
| | - Shinichi Sato
- Department of Dermatology, Faculty of Medicine, University of Tokyo, Japan.
| |
Collapse
|