1
|
Smirnov S, Mateikovich P, Samochernykh K, Shlyakhto E. Recent advances on CAR-T signaling pave the way for prolonged persistence and new modalities in clinic. Front Immunol 2024; 15:1335424. [PMID: 38455066 PMCID: PMC10918004 DOI: 10.3389/fimmu.2024.1335424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment of hematological malignancies. The importance of the receptor costimulatory domain for long-term CAR-T cell engraftment and therapeutic efficacy was demonstrated with second-generation CAR-T cells. Fifth generation CAR-T cells are currently in preclinical trials. At the same time, the processes that orchestrate the activation and differentiation of CAR-T cells into a specific phenotype that predisposes them to long-term persistence are not fully understood. This review highlights ongoing research aimed at elucidating the role of CAR domains and T-cell signaling molecules involved in these processes.
Collapse
Affiliation(s)
- Sergei Smirnov
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia
| | - Polina Mateikovich
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia
| | - Konstantin Samochernykh
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia
| | - Evgeny Shlyakhto
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia
| |
Collapse
|
2
|
Cassioli C, Patrussi L, Valitutti S, Baldari CT. Learning from TCR Signaling and Immunological Synapse Assembly to Build New Chimeric Antigen Receptors (CARs). Int J Mol Sci 2022; 23:14255. [PMID: 36430728 PMCID: PMC9694822 DOI: 10.3390/ijms232214255] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell immunotherapy is a revolutionary pillar in cancer treatment. Clinical experience has shown remarkable successes in the treatment of certain hematological malignancies but only limited efficacy against B cell chronic lymphocytic leukemia (CLL) and other cancer types, especially solid tumors. A wide range of engineering strategies have been employed to overcome the limitations of CAR T cell therapy. However, it has become increasingly clear that CARs have unique, unexpected features; hence, a deep understanding of how CARs signal and trigger the formation of a non-conventional immunological synapse (IS), the signaling platform required for T cell activation and execution of effector functions, would lead a shift from empirical testing to the rational design of new CAR constructs. Here, we review current knowledge of CARs, focusing on their structure, signaling and role in CAR T cell IS assembly. We, moreover, discuss the molecular features accounting for poor responses in CLL patients treated with anti-CD19 CAR T cells and propose CLL as a paradigm for diseases connected to IS dysfunctions that could significantly benefit from the development of novel CARs to generate a productive anti-tumor response.
Collapse
Affiliation(s)
- Chiara Cassioli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Laura Patrussi
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Salvatore Valitutti
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31037 Toulouse, France
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France
| | - Cosima T. Baldari
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| |
Collapse
|
3
|
Liu C, Ma L, Wang Y, Zhao J, Chen P, Chen X, Wang Y, Hu Y, Liu Y, Jia X, Yang Z, Yin X, Wu J, Wu S, Zheng H, Ma X, Sun X, He Y, Lin L, Fu Y, Liao K, Zhou X, Jiang S, Fu G, Tang J, Han W, Chen XL, Fan W, Hong Y, Han J, Huang X, Li BA, Xiao N, Xiao C, Fu G, Liu WH. Glycogen synthase kinase 3 drives thymocyte egress by suppressing β-catenin activation of Akt. SCIENCE ADVANCES 2021; 7:eabg6262. [PMID: 34623920 PMCID: PMC8500522 DOI: 10.1126/sciadv.abg6262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
Molecular pathways controlling emigration of mature thymocytes from thymus to the periphery remain incompletely understood. Here, we show that T cell–specific ablation of glycogen synthase kinase 3 (GSK3) led to severely impaired thymic egress. In the absence of GSK3, β-catenin accumulated in the cytoplasm, where it associated with and activated Akt, leading to phosphorylation and degradation of Foxo1 and downregulation of Klf2 and S1P1 expression, thereby preventing emigration of thymocytes. A cytoplasmic membrane-localized β-catenin excluded from the nucleus promoted Akt activation, suggesting a new function of β-catenin independent of its role as a transcriptional activator. Furthermore, genetic ablation of β-catenin, retroviral expression of a dominant negative Akt mutant, and transgenic expression of a constitutively active Foxo1 restored emigration of GSK3-deficient thymocytes. Our findings establish an essential role for GSK3 in thymocyte egress and reveal a previously unidentified signaling function of β-catenin in the cytoplasm.
Collapse
Affiliation(s)
- Chenfeng Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Lei Ma
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yuxuan Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiayi Zhao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Pengda Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xian Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yingxin Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yanyan Hu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yun Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xian Jia
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhanghua Yang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xingzhi Yin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Suqin Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Haiping Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaohong Ma
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiufeng Sun
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Ying He
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Lianghua Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yubing Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kunyu Liao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaojuan Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shan Jiang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Guofeng Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jian Tang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wei Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiao Lei Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wenzhu Fan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yazhen Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiangyang Huang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Bo-An Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Nengming Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
4
|
Machiyama H, Yamaguchi T, Watanabe TM, Yanagida T, Fujita H. Activation probability of a single naïve T cell upon TCR ligation is controlled by T cells interacting with the same antigen-presenting cell. FEBS Lett 2021; 595:1512-1524. [PMID: 33826750 DOI: 10.1002/1873-3468.14082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 11/07/2022]
Abstract
Accurate recognition of antigens by specific T cells is crucial for adaptive immunity to work properly. The activation of a T-cell antigen-specific response by an antigen-presenting cell (APC) has not been clearly measured at a single T-cell level. It is also unknown whether the cell-extrinsic environment alters antigen recognition by a T cell. To measure the activation probability of a single T cell by an APC, we performed a single-cell live imaging assay and found that the activation probability changes depending not only on the antigens but also on the interactions of other T cells with the APC. We found that the specific reactivity of single naïve T cells was poor. However, their antigen-specific reactivity increased drastically when attached to an APC interacting with activated T cells. Activation of T cells was suppressed when regulatory T cells interacted with the APC. These findings suggest that although the ability of APCs to activate an antigen-specific naïve T cell is low at a single-cell level, the surrounding environment of APCs improves the specificity of the bulk response.
Collapse
Affiliation(s)
- Hiroaki Machiyama
- Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Immunology, Tokyo Medical University, Japan
| | - Tomoyuki Yamaguchi
- Immunology Frontier Research Center, Osaka University, Suita, Japan.,Research Institute, Nozaki Tokushukai Hospital, Daito, Japan
| | - Tomonobu M Watanabe
- RIKEN Center for Biosystems Dynamics Research, Laboratory for Comprehensive Bioimaging, Kobe, Japan.,Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Japan
| | - Toshio Yanagida
- Immunology Frontier Research Center, Osaka University, Suita, Japan.,RIKEN Center for Biosystems Dynamics Research, Laboratory for Comprehensive Bioimaging, Kobe, Japan.,Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita, Japan
| | - Hideaki Fujita
- Immunology Frontier Research Center, Osaka University, Suita, Japan.,RIKEN Center for Biosystems Dynamics Research, Laboratory for Comprehensive Bioimaging, Kobe, Japan.,Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Japan
| |
Collapse
|
5
|
Connolly A, Panes R, Tual M, Lafortune R, Bellemare-Pelletier A, Gagnon E. TMEM16F mediates bystander TCR-CD3 membrane dissociation at the immunological synapse and potentiates T cell activation. Sci Signal 2021; 14:eabb5146. [PMID: 33758060 DOI: 10.1126/scisignal.abb5146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Electrostatic interactions regulate many aspects of T cell receptor (TCR) activity, including enabling the dynamic binding of the TCR-associated CD3ε and CD3ζ chains to anionic lipids in the plasma membrane to prevent spontaneous phosphorylation. Substantial changes in the electrostatic potential of the plasma membrane occur at the immunological synapse, the interface between a T cell and an antigen-presenting cell. Here, we investigated how the electrostatic interactions that promote dynamic membrane binding of the TCR-CD3 cytoplasmic domains are modulated during signaling and affect T cell activation. We found that Ca2+-dependent activation of the phosphatidylserine scramblase TMEM16F, which was previously implicated in T cell activation, reduced the electrostatic potential of the plasma membrane during immunological synapse formation by locally redistributing phosphatidylserine. This, in turn, increased the dissociation of bystander TCR-CD3 cytoplasmic domains from the plasma membrane and enhanced TCR-dependent signaling and consequently T cell activation. This study establishes the molecular basis for the role of TMEM16F in bystander TCR-induced signal amplification and identifies enhancement of TMEM16F function as a potential therapeutic strategy for promoting T cell activation.
Collapse
Affiliation(s)
- Audrey Connolly
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| | - Rébecca Panes
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| | - Margaux Tual
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| | - Raphaël Lafortune
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| | - Angélique Bellemare-Pelletier
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada
| | - Etienne Gagnon
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada.
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| |
Collapse
|
6
|
Descamps D, Evnouchidou I, Caillens V, Drajac C, Riffault S, van Endert P, Saveanu L. The Role of Insulin Regulated Aminopeptidase in Endocytic Trafficking and Receptor Signaling in Immune Cells. Front Mol Biosci 2020; 7:583556. [PMID: 33195428 PMCID: PMC7606930 DOI: 10.3389/fmolb.2020.583556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Insulin regulated aminopeptidase (IRAP) is a type II transmembrane protein with broad tissue distribution initially identified as a major component of Glut4 storage vesicles (GSV) in adipocytes. Despite its almost ubiquitous expression, IRAP had been extensively studied mainly in insulin responsive cells, such as adipocytes and muscle cells. In these cells, the enzyme displays a complex intracellular trafficking pattern regulated by insulin. Early studies using fusion proteins joining the IRAP cytosolic domain to various reporter proteins, such as GFP or the transferrin receptor (TfR), showed that the complex and regulated trafficking of the protein depends on its cytosolic domain. This domain contains several motifs involved in IRAP trafficking, as demonstrated by mutagenesis studies. Also, proteomic studies and yeast two-hybrid experiments showed that the IRAP cytosolic domain engages in multiple protein interactions with cytoskeleton components and vesicular trafficking adaptors. These findings led to the hypothesis that IRAP is not only a cargo of GSV but might be a part of the sorting machinery that controls GSV dynamics. Recent work in adipocytes, immune cells, and neurons confirmed this hypothesis and demonstrated that IRAP has a dual function. Its carboxy-terminal domain located inside endosomes is responsible for the aminopeptidase activity of the enzyme, while its amino-terminal domain located in the cytosol functions as an endosomal trafficking adaptor. In this review, we recapitulate the published protein interactions of IRAP and summarize the increasing body of evidence indicating that IRAP plays a role in intracellular trafficking of several proteins. We describe the impact of IRAP deletion or depletion on endocytic trafficking and the consequences on immune cell functions. These include the ability of dendritic cells to cross-present antigens and prime adaptive immune responses, as well as the control of innate and adaptive immune receptor signaling and modulation of inflammatory responses.
Collapse
Affiliation(s)
| | - Irini Evnouchidou
- Université de Paris, Centre de recherche sur l'inflammation, INSERM U1149, CNRS ERL8252, Paris, France.,Inovarion, Paris, France
| | - Vivien Caillens
- Université de Paris, Centre de recherche sur l'inflammation, INSERM U1149, CNRS ERL8252, Paris, France
| | - Carole Drajac
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jou-en-Josas, France
| | - Sabine Riffault
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jou-en-Josas, France
| | - Peter van Endert
- Université de Paris, Centre de recherche sur l'inflammation, INSERM U1149, CNRS ERL8252, Paris, France.,Université de Paris, INSERM Unité 1151, CNRS UMR 8253, Paris, France.,Service d'immunologie biologique, AP-HP, Hôpital Necker, Paris, France
| | - Loredana Saveanu
- Université de Paris, Centre de recherche sur l'inflammation, INSERM U1149, CNRS ERL8252, Paris, France
| |
Collapse
|
7
|
Baldin AV, Savvateeva LV, Bazhin AV, Zamyatnin AA. Dendritic Cells in Anticancer Vaccination: Rationale for Ex Vivo Loading or In Vivo Targeting. Cancers (Basel) 2020; 12:cancers12030590. [PMID: 32150821 PMCID: PMC7139354 DOI: 10.3390/cancers12030590] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) have shown great potential as a component or target in the landscape of cancer immunotherapy. Different in vivo and ex vivo strategies of DC vaccine generation with different outcomes have been proposed. Numerous clinical trials have demonstrated their efficacy and safety in cancer patients. However, there is no consensus regarding which DC-based vaccine generation method is preferable. A problem of result comparison between trials in which different DC-loading or -targeting approaches have been applied remains. The employment of different DC generation and maturation methods, antigens and administration routes from trial to trial also limits the objective comparison of DC vaccines. In the present review, we discuss different methods of DC vaccine generation. We conclude that standardized trial designs, treatment settings and outcome assessment criteria will help to determine which DC vaccine generation approach should be applied in certain cancer cases. This will result in a reduction in alternatives in the selection of preferable DC-based vaccine tactics in patient. Moreover, it has become clear that the application of a DC vaccine alone is not sufficient and combination immunotherapy with recent advances, such as immune checkpoint inhibitors, should be employed to achieve a better clinical response and outcome.
Collapse
Affiliation(s)
- Alexey V. Baldin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Lyudmila V. Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians University of Munich, 81377 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
- Belozersky Institute of Physico-Chemical Biology, Department of Cell Signaling, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +74-956-229-843
| |
Collapse
|
8
|
Finetti F, Capitani N, Baldari CT. Emerging Roles of the Intraflagellar Transport System in the Orchestration of Cellular Degradation Pathways. Front Cell Dev Biol 2019; 7:292. [PMID: 31803744 PMCID: PMC6877659 DOI: 10.3389/fcell.2019.00292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/06/2019] [Indexed: 12/27/2022] Open
Abstract
Ciliated cells exploit a specific transport system, the intraflagellar transport (IFT) system, to ensure the traffic of molecules from the cell body to the cilium. However, it is now clear that IFT activity is not restricted to cilia-related functions. This is strikingly exemplified by the observation that IFT proteins play important roles in cells lacking a primary cilium, such as lymphocytes. Indeed, in T cells the IFT system regulates the polarized transport of endosome-associated T cell antigen receptors and signaling mediators during assembly of the immune synapse, a specialized interface that forms on encounter with a cognate antigen presenting cell and on which T cell activation and effector function crucially depend. Cellular degradation pathways have recently emerged as new extraciliary functions of the IFT system. IFT proteins have been demonstrated to regulate autophagy in ciliated cells through their ability to recruit the autophagy machinery to the base of the cilium. We have now implicated the IFT component IFT20 in another central degradation process that also controls the latest steps in autophagy, namely lysosome function, by regulating the cation-independent mannose-6-phosphate receptor (CI-MPR)-dependent lysosomal targeting of acid hydrolases. This involves the ability of IFT20 to act as an adaptor coupling the CI-MPR to dynein for retrograde transport to the trans-Golgi network. In this short review we will discuss the emerging roles of IFT proteins in cellular degradation pathways.
Collapse
Affiliation(s)
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|
9
|
Horwitz DA, Fahmy TM, Piccirillo CA, La Cava A. Rebalancing Immune Homeostasis to Treat Autoimmune Diseases. Trends Immunol 2019; 40:888-908. [PMID: 31601519 DOI: 10.1016/j.it.2019.08.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 08/01/2019] [Accepted: 08/14/2019] [Indexed: 12/18/2022]
Abstract
During homeostasis, interactions between tolerogenic dendritic cells (DCs), self-reactive T cells, and T regulatory cells (Tregs) contribute to maintaining mammalian immune tolerance. In response to infection, immunogenic DCs promote the generation of proinflammatory effector T cell subsets. When complex homeostatic mechanisms maintaining the balance between regulatory and effector functions become impaired, autoimmune diseases can develop. We discuss some of the newest advances on the mechanisms of physiopathologic homeostasis that can be employed to develop strategies to restore a dysregulated immune equilibrium. Some of these designs are based on selectively activating regulators of immunity and inflammation instead of broadly suppressing these processes. Promising approaches include the use of nanoparticles (NPs) to restore Treg control over self-reactive cells, aiming to achieve long-term disease remission, and potentially to prevent autoimmunity in susceptible individuals.
Collapse
Affiliation(s)
- David A Horwitz
- General Nanotherapeutics, LLC, Santa Monica, CA, USA; Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Tarek M Fahmy
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, CT, USA; Chemical and Environmental Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, CT, USA; Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada; Program in Infectious Disease and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montréal, QC, Canada; Centre of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Antonio La Cava
- Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
10
|
Fang J, Muto T, Kleppe M, Bolanos LC, Hueneman KM, Walker CS, Sampson L, Wellendorf AM, Chetal K, Choi K, Salomonis N, Choi Y, Zheng Y, Cancelas JA, Levine RL, Starczynowski DT. TRAF6 Mediates Basal Activation of NF-κB Necessary for Hematopoietic Stem Cell Homeostasis. Cell Rep 2019; 22:1250-1262. [PMID: 29386112 PMCID: PMC5971064 DOI: 10.1016/j.celrep.2018.01.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/14/2017] [Accepted: 01/04/2018] [Indexed: 11/03/2022] Open
Abstract
Basal nuclear factor κB (NF-κB) activation is required for hematopoietic stem cell (HSC) homeostasis in the absence of inflammation; however, the upstream mediators of basal NF-κB signaling are less well understood. Here, we describe TRAF6 as an essential regulator of HSC homeostasis through basal activation of NF-κB. Hematopoietic-specific deletion of Traf6 resulted in impaired HSC self-renewal and fitness. Gene expression, RNA splicing, and molecular analyses of Traf6-deficient hematopoietic stem/progenitor cells (HSPCs) revealed changes in adaptive immune signaling, innate immune signaling, and NF-κB signaling, indicating that signaling via TRAF6 in the absence of cytokine stimulation and/or infection is required for HSC function. In addition, we established that loss of IκB kinase beta (IKKβ)-mediated NF-κB activation is responsible for the major hematopoietic defects observed in Traf6-deficient HSPC as deletion of IKKβ similarly resulted in impaired HSC self-renewal and fitness. Taken together, TRAF6 is required for HSC homeostasis by maintaining a minimal threshold level of IKKβ/NF-κB signaling.
Collapse
Affiliation(s)
- Jing Fang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tomoya Muto
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Maria Kleppe
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lyndsey C Bolanos
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kathleen M Hueneman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Callum S Walker
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Leesa Sampson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ashley M Wellendorf
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jose A Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
11
|
Albert S, Koristka S, Gerbaulet A, Cartellieri M, Arndt C, Feldmann A, Berndt N, Loureiro LR, von Bonin M, Ehninger G, Eugster A, Bonifacio E, Bornhäuser M, Bachmann MP, Ehninger A. Tonic Signaling and Its Effects on Lymphopoiesis of CAR-Armed Hematopoietic Stem and Progenitor Cells. THE JOURNAL OF IMMUNOLOGY 2019; 202:1735-1746. [PMID: 30728213 DOI: 10.4049/jimmunol.1801004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/04/2019] [Indexed: 01/01/2023]
Abstract
Long-term survival of adoptively transferred chimeric Ag receptor (CAR) T cells is often limited. Transplantation of hematopoietic stem cells (HSCs) transduced to express CARs could help to overcome this problem as CAR-armed HSCs can continuously deliver CAR+ multicell lineages (e.g., T cells, NK cells). In dependence on the CAR construct, a variable extent of tonic signaling in CAR T cells was reported; thus, effects of CAR-mediated tonic signaling on the hematopoiesis of CAR-armed HSCs is unclear. To assess the effects of tonic signaling, two CAR constructs were established and analyzed 1) a signaling CAR inducing a solid Ag-independent tonic signaling termed CAR-28/ζ and 2) a nonstimulating control CAR construct lacking intracellular signaling domains termed CAR-Stop. Bone marrow cells from immunocompetent mice were isolated, purified for HSC-containing Lin-cKit+ cells or the Lin-cKit+ Sca-1+ subpopulation (Lin-Sca-1+cKit+), and transduced with both CAR constructs. Subsequently, modified bone marrow cells were transferred into irradiated mice, in which they successfully engrafted and differentiated into hematopoietic progenitors. HSCs expressing the CAR-Stop sustained normal hematopoiesis. In contrast, expression of the CAR-28/ζ led to elimination of mature CAR+ T and B cells, suggesting that the CAR-mediated tonic signaling mimics autorecognition via the newly recombined immune receptors in the developing lymphocytes.
Collapse
Affiliation(s)
- Susann Albert
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.,University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Stefanie Koristka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Alexander Gerbaulet
- Institute of Immunology, Medical Faculty Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | | | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Nicole Berndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Liliana R Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Malte von Bonin
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Gerhard Ehninger
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Anne Eugster
- Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, 01307 Dresden, Germany; and
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, 01307 Dresden, Germany; and
| | - Martin Bornhäuser
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Michael P Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; .,University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Armin Ehninger
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,GEMoaB Monoclonals GmbH, 01307 Dresden, Germany
| |
Collapse
|
12
|
Ajina A, Maher J. Strategies to Address Chimeric Antigen Receptor Tonic Signaling. Mol Cancer Ther 2018; 17:1795-1815. [PMID: 30181329 PMCID: PMC6130819 DOI: 10.1158/1535-7163.mct-17-1097] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/19/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022]
Abstract
Adoptive cell transfer using chimeric antigen receptors (CAR) has emerged as one of the most promising new therapeutic modalities for patients with relapsed or refractory B-cell malignancies. Thus far, results in patients with advanced solid tumors have proven disappointing. Constitutive tonic signaling in the absence of ligand is an increasingly recognized complication when deploying these synthetic fusion receptors and can be a cause of poor antitumor efficacy, impaired survival, and reduced persistence in vivo In parallel, ligand-dependent tonic signaling can mediate toxicity and promote T-cell anergy, exhaustion, and activation-induced cell death. Here, we review the mechanisms underpinning CAR tonic signaling and highlight the wide variety of effects that can emerge after making subtle structural changes or altering the methodology of CAR transduction. We highlight strategies to prevent unconstrained tonic signaling and address its deleterious consequences. We also frame this phenomenon in the context of endogenous TCR tonic signaling, which has been shown to regulate peripheral tolerance, facilitate the targeting of foreign antigens, and suggest opportunities to coopt ligand-dependent CAR tonic signaling to facilitate in vivo persistence and efficacy. Mol Cancer Ther; 17(9); 1795-815. ©2018 AACR.
Collapse
MESH Headings
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Adam Ajina
- CAR Mechanics Group, King's College London, London, United Kingdom.
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
| | - John Maher
- CAR Mechanics Group, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Department of Immunology, Eastbourne Hospital, East Sussex, United Kingdom
| |
Collapse
|
13
|
Stress-testing the relationship between T cell receptor/peptide-MHC affinity and cross-reactivity using peptide velcro. Proc Natl Acad Sci U S A 2018; 115:E7369-E7378. [PMID: 30021852 DOI: 10.1073/pnas.1802746115] [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] [Indexed: 01/01/2023] Open
Abstract
T cell receptors (TCRs) bind to peptide-major histocompatibility complex (pMHC) with low affinity (Kd ∼ μM), which is generally assumed to facilitate cross-reactive TCR "scanning" of ligands. To understand the relationship between TCR/pMHC affinity and cross-reactivity, we sought to engineer an additional weak interaction, termed "velcro," between the TCR and pMHC to probe the specificities of TCRs at relatively low and high affinities. This additional interaction was generated through an eight-amino acid peptide library covalently linked to the N terminus of the MHC-bound peptide. Velcro was selected through an affinity-based isolation and was subsequently shown to enhance the cognate TCR/pMHC affinity in a peptide-dependent manner by ∼10-fold. This was sufficient to convert a nonstimulatory ultra-low-affinity ligand into a stimulatory ligand. An X-ray crystallographic structure revealed how velcro interacts with the TCR. To probe TCR cross-reactivity, we screened TCRs against yeast-displayed pMHC libraries with and without velcro, and found that the peptide cross-reactivity profiles of low-affinity (Kd > 100 μM) and high-affinity (Kd ∼ μM) TCR/pMHC interactions are remarkably similar. The conservation of recognition of the TCR for pMHC across affinities reveals the nature of low-affinity ligands for which there are important biological functions and has implications for understanding the specificities of affinity-matured TCRs.
Collapse
|
14
|
Stead SO, McInnes SJP, Kireta S, Rose PD, Jesudason S, Rojas-Canales D, Warther D, Cunin F, Durand JO, Drogemuller CJ, Carroll RP, Coates PT, Voelcker NH. Manipulating human dendritic cell phenotype and function with targeted porous silicon nanoparticles. Biomaterials 2017; 155:92-102. [PMID: 29175084 DOI: 10.1016/j.biomaterials.2017.11.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/13/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DC) are the most potent antigen-presenting cells and are fundamental for the establishment of transplant tolerance. The Dendritic Cell-Specific Intracellular adhesion molecule-3-Grabbing Non-integrin (DC-SIGN; CD209) receptor provides a target for dendritic cell therapy. Biodegradable and high-surface area porous silicon (pSi) nanoparticles displaying anti-DC-SIGN antibodies and loaded with the immunosuppressant rapamycin (Sirolimus) serve as a fit-for-purpose platform to target and modify DC. Here, we describe the fabrication of rapamycin-loaded DC-SIGN displaying pSi nanoparticles, the uptake efficiency into DC and the extent of nanoparticle-induced modulation of phenotype and function. DC-SIGN antibody displaying pSi nanoparticles favourably targeted and were phagocytosed by monocyte-derived and myeloid DC in whole human blood in a time- and dose-dependent manner. DC preconditioning with rapamycin-loaded nanoparticles, resulted in a maturation resistant phenotype and significantly suppressed allogeneic T-cell proliferation.
Collapse
Affiliation(s)
| | - Steven J P McInnes
- Future Industries Institute, University of South Australia, Adelaide, Australia
| | - Svjetlana Kireta
- Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - Peter D Rose
- University of Adelaide, Department of Medicine, Adelaide, Australia
| | - Shilpanjali Jesudason
- University of Adelaide, Department of Medicine, Adelaide, Australia; Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - Darling Rojas-Canales
- Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - David Warther
- Institut Charles Gerhardt Montpellier, UMR 5253 CNRS -ENSCM-UM2-UM1, Ecole Nationale Supérieure de Chimie de Montpellier, 34296, Montpellier, France
| | - Frédérique Cunin
- Institut Charles Gerhardt Montpellier, UMR 5253 CNRS -ENSCM-UM2-UM1, Ecole Nationale Supérieure de Chimie de Montpellier, 34296, Montpellier, France
| | - Jean-Olivier Durand
- Institut Charles Gerhardt Montpellier, UMR 5253 CNRS -ENSCM-UM2-UM1, Ecole Nationale Supérieure de Chimie de Montpellier, 34296, Montpellier, France
| | - Christopher J Drogemuller
- University of Adelaide, Department of Medicine, Adelaide, Australia; Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - Robert P Carroll
- University of Adelaide, Department of Medicine, Adelaide, Australia; Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - P Toby Coates
- University of Adelaide, Department of Medicine, Adelaide, Australia; Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia.
| | - Nicolas H Voelcker
- Future Industries Institute, University of South Australia, Adelaide, Australia; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, Australia; Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia; Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
15
|
A complex of Neuroplastin and Plasma Membrane Ca 2+ ATPase controls T cell activation. Sci Rep 2017; 7:8358. [PMID: 28827723 PMCID: PMC5566957 DOI: 10.1038/s41598-017-08519-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/10/2017] [Indexed: 12/24/2022] Open
Abstract
The outcome of T cell activation is determined by mechanisms that balance Ca2+ influx and clearance. Here we report that murine CD4 T cells lacking Neuroplastin (Nptn -/-), an immunoglobulin superfamily protein, display elevated cytosolic Ca2+ and impaired post-stimulation Ca2+ clearance, along with increased nuclear levels of NFAT transcription factor and enhanced T cell receptor-induced cytokine production. On the molecular level, we identified plasma membrane Ca2+ ATPases (PMCAs) as the main interaction partners of Neuroplastin. PMCA levels were reduced by over 70% in Nptn -/- T cells, suggesting an explanation for altered Ca2+ handling. Supporting this, Ca2+ extrusion was impaired while Ca2+ levels in internal stores were increased. T cells heterozygous for PMCA1 mimicked the phenotype of Nptn -/- T cells. Consistent with sustained Ca2+ levels, differentiation of Nptn -/- T helper cells was biased towards the Th1 versus Th2 subset. Our study thus establishes Neuroplastin-PMCA modules as important regulators of T cell activation.
Collapse
|
16
|
Waisman A, Lukas D, Clausen BE, Yogev N. Dendritic cells as gatekeepers of tolerance. Semin Immunopathol 2017; 39:153-163. [PMID: 27456849 DOI: 10.1007/s00281-016-0583-z] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/07/2016] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DC) are unique hematopoietic cells, linking innate and adaptive immune responses. In particular, they are considered as the most potent antigen presenting cells, governing both T cell immunity and tolerance. In view of their exceptional ability to present antigen and to interact with T cells, DC play distinct roles in shaping T cell development, differentiation and function. The outcome of the DC-T cell interaction is determined by the state of DC maturation, the type of DC subset, the cytokine microenvironment and the tissue location. Both regulatory T cells (Tregs) and DC are indispensable for maintaining central and peripheral tolerance. Over the past decade, accumulating data indicate that DC critically contribute to Treg differentiation and homeostasis.
Collapse
Affiliation(s)
- Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Dominika Lukas
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Microbiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nir Yogev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
17
|
Guedj C, Abraham N, Jullié D, Randriamampita C. T cell adhesion triggers an early signaling pole distal to the immune synapse. J Cell Sci 2016; 129:2526-37. [PMID: 27185862 DOI: 10.1242/jcs.182311] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 05/09/2016] [Indexed: 01/09/2023] Open
Abstract
The immunological synapse forms at the interface between a T cell and an antigen-presenting cell after foreign antigen recognition. The immunological synapse is considered to be the site where the signaling cascade leading to T lymphocyte activation is triggered. Here, we show that another signaling region can be detected before formation of the synapse at the opposite pole of the T cell. This structure appears during the first minute after the contact forms, is transient and contains all the classic components that have been previously described at the immunological synapse. Its formation is independent of antigen recognition but is driven by adhesion itself. It constitutes a reservoir of signaling molecules that are potentially ready to be sent to the immunological synapse through a microtubule-dependent pathway. The antisynapse can thus be considered as a pre-synapse that is triggered independently of antigen recognition.
Collapse
Affiliation(s)
- Chloé Guedj
- INSERM, U1016, Institut Cochin, Infection, Immunity and Inflammation Department, 22 rue Méćhain, Paris 75014, France CNRS, UMR8104, Paris 75014, France Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Nicolas Abraham
- INSERM, U1016, Institut Cochin, Infection, Immunity and Inflammation Department, 22 rue Méćhain, Paris 75014, France CNRS, UMR8104, Paris 75014, France Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Damien Jullié
- INSERM, U1016, Institut Cochin, Infection, Immunity and Inflammation Department, 22 rue Méćhain, Paris 75014, France CNRS, UMR8104, Paris 75014, France Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Clotilde Randriamampita
- INSERM, U1016, Institut Cochin, Infection, Immunity and Inflammation Department, 22 rue Méćhain, Paris 75014, France CNRS, UMR8104, Paris 75014, France Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| |
Collapse
|
18
|
High-density preculture of PBMCs restores defective sensitivity of circulating CD8 T cells to virus- and tumor-derived antigens. Blood 2015; 126:185-94. [DOI: 10.1182/blood-2015-01-622704] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/27/2015] [Indexed: 01/13/2023] Open
Abstract
Key Points
CD8 memory T cells in PBMCs are antigen-hyporesponsive due to loss of priming by tissue-dependent interactions. Preculture at high cell density allows the detection of antiviral and antitumor responses that may be overlooked without this step.
Collapse
|
19
|
Immunotherapy for lung cancer: for whom the bell tolls? Tumour Biol 2015; 36:1411-22. [PMID: 25736929 DOI: 10.1007/s13277-015-3285-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 02/18/2015] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death and accounts for approximately 30% of all cancer deaths. Despite the recent developments in personalized therapy, the prognosis in lung cancer is still very poor. Immunotherapy is now emerging as a new hope for patients with lung cancer. It is well known that standard chemotherapeutic regimens have devastating effects for the patient's immune system. Therefore, the aim of immunotherapy is to specifically enhance the immune response against the tumour. Recently, many trials addressed the role of such therapies for metastatic non-small cell lung cancer (NSCLC) treatment: ipilimumab, tremelimumab, nivolumab and pembrolizumab are immunotherapeutic agents of high relevance in this field. Anti-tumour vaccines, as well as dendritic cell-based therapies, have emerged as potent inducers of immune response against the tumour. Herein, we will review some of the most promising cancer immunotherapies, highlighting their advantages and try to understand, in an immunological perspective, the missteps associated with the current treatments for cancer.
Collapse
|
20
|
Abstract
Dynamic tuning of cellular responsiveness as a result of repeated stimuli improves the ability of cells to distinguish physiologically meaningful signals from each other and from noise. In particular, lymphocyte activation thresholds are subject to tuning, which contributes to maintaining tolerance to self-antigens and persisting foreign antigens, averting autoimmunity and immune pathogenesis, but allowing responses to strong, structured perturbations that are typically associated with acute infection. Such tuning is also implicated in conferring flexibility to positive selection in the thymus, in controlling the magnitude of the immune response, and in generating memory cells. Additional functional properties are dynamically and differentially tuned in parallel via subthreshold contact interactions between developing or mature lymphocytes and self-antigen-presenting cells. These interactions facilitate and regulate lymphocyte viability, maintain their functional integrity, and influence their responses to foreign antigens and accessory signals, qualitatively and quantitatively. Bidirectional tuning of T cells and antigen-presenting cells leads to the definition of homeostatic set points, thus maximizing clonal diversity.
Collapse
Affiliation(s)
- Zvi Grossman
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; ,
| | | |
Collapse
|
21
|
Knolle PA, Böttcher J, Huang LR. The role of hepatic immune regulation in systemic immunity to viral infection. Med Microbiol Immunol 2014; 204:21-7. [PMID: 25523194 DOI: 10.1007/s00430-014-0371-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 10/27/2014] [Indexed: 12/19/2022]
Abstract
The liver has particular immune functions attributed by its unique microenvironment and its liver-resident cell populations. During autoimmunity and viral hepatitis, the liver serves as target for effector responses of immune cells. However, skewing of effector T cell functions through tolerogenic liver-resident antigen-presenting cells and through the immune regulatory hepatic microenvironment. Importantly, the liver also participates in shaping systemic antigen-specific immunity. Local antigen-presenting cell populations, in particular liver sinusoidal endothelial cells (LSECs), cross-present soluble, circulating or hepatocyte-derived antigens to naïve CD8 T cells. Upon priming by cross-presenting LSECs, naïve CD8 T cells develop into a unique population of antigen-experienced memory-like T cell population that can be reactivated in an inflammatory context to protect against infection with viruses or bacteria. Furthermore, upon prolonged inflammatory TNF-dependent signaling, the induction of intrahepatic myeloid cell aggregates for T cell population expansion (iMATEs) is observed in liver tissue. iMATEs are formed by inflammatory monocytes developing into dendritic cells and function to attract recently activated CD8 T cells. Those CD8 T cells located within the cocoon-like iMATE structure show strong proliferation initiated by co-stimulatory signaling. Locally expanded CD8 T cells are key to control acute and chronic viral infections. The mechanistic understanding of local hepatic T cell priming and local expansion of effector CD8 T cells will help to develop novel therapeutic vaccination strategies.
Collapse
Affiliation(s)
- Percy A Knolle
- Institute of Molecular Immunology, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, 81675, Munich, Germany,
| | | | | |
Collapse
|
22
|
Khailaie S, Robert PA, Toker A, Huehn J, Meyer-Hermann M. A signal integration model of thymic selection and natural regulatory T cell commitment. THE JOURNAL OF IMMUNOLOGY 2014; 193:5983-96. [PMID: 25392533 DOI: 10.4049/jimmunol.1400889] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The extent of TCR self-reactivity is the basis for selection of a functional and self-tolerant T cell repertoire and is quantified by repeated engagement of TCRs with a diverse pool of self-peptides complexed with self-MHC molecules. The strength of a TCR signal depends on the binding properties of a TCR to the peptide and the MHC, but it is not clear how the specificity to both components drives fate decisions. In this study, we propose a TCR signal-integration model of thymic selection that describes how thymocytes decide among distinct fates, not only based on a single TCR-ligand interaction, but taking into account the TCR stimulation history. These fates are separated based on sustained accumulated signals for positive selection and transient peak signals for negative selection. This spans up the cells into a two-dimensional space where they are either neglected, positively selected, negatively selected, or selected as natural regulatory T cells (nTregs). We show that the dynamics of the integrated signal can serve as a successful basis for extracting specificity of thymocytes to MHC and detecting the existence of cognate self-peptide-MHC. It allows to select a self-MHC-biased and self-peptide-tolerant T cell repertoire. Furthermore, nTregs in the model are enriched with MHC-specific TCRs. This allows nTregs to be more sensitive to activation and more cross-reactive than conventional T cells. This study provides a mechanistic model showing that time integration of TCR-mediated signals, as opposed to single-cell interaction events, is needed to gain a full view on the properties emerging from thymic selection.
Collapse
Affiliation(s)
- Sahamoddin Khailaie
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Philippe A Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique, 34293 Montpellier, France
| | - Aras Toker
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; and
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; and
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Institute for Biochemistry, Biotechnology, and Bioinformatics, University of Technology Braunschweig, 38106 Braunschweig, Germany
| |
Collapse
|
23
|
Delpoux A, Yakonowsky P, Durand A, Charvet C, Valente M, Pommier A, Bonilla N, Martin B, Auffray C, Lucas B. TCR signaling events are required for maintaining CD4 regulatory T cell numbers and suppressive capacities in the periphery. THE JOURNAL OF IMMUNOLOGY 2014; 193:5914-23. [PMID: 25381435 DOI: 10.4049/jimmunol.1400477] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CD4 regulatory T cells (Tregs) can be subdivided into two subsets according to Ly-6C expression in the periphery. Phenotypic analysis, imaging, and adoptive-transfer experiments of peripheral Ly-6C(-) and Ly-6C(+) Tregs reveal that the nonexpression of Ly-6C by ∼70% of peripheral Tregs depends on TCR signaling events. Interestingly, Ly-6C(-) Tregs express higher surface amounts of key immunosuppressive molecules than do Ly-6C(+) Tregs and produce constitutively anti-inflammatory cytokines. In line with their phenotype, Ly-6C(+) Tregs exhibit poor suppressive capacities in vitro and in vivo. Finally, although Ly-6C(-) Tregs maintain their numbers with age, Ly-6C(+) Tregs gradually disappear. Altogether, our data strongly suggest that both the survival and suppressive functions of peripheral CD4 Tregs rely on their ability to receive strong TCR signals.
Collapse
Affiliation(s)
- Arnaud Delpoux
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Philippe Yakonowsky
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Aurélie Durand
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Céline Charvet
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Michael Valente
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Arnaud Pommier
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Nelly Bonilla
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Bruno Martin
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Cédric Auffray
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Bruno Lucas
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, 75014 Paris, France
| |
Collapse
|
24
|
Martin SF. Adaptation in the innate immune system and heterologous innate immunity. Cell Mol Life Sci 2014; 71:4115-30. [PMID: 24997561 PMCID: PMC11113124 DOI: 10.1007/s00018-014-1676-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/18/2014] [Accepted: 06/30/2014] [Indexed: 01/05/2023]
Abstract
The innate immune system recognizes deviation from homeostasis caused by infectious or non-infectious assaults. The threshold for its activation seems to be established by a calibration process that includes sensing of microbial molecular patterns from commensal bacteria and of endogenous signals. It is becoming increasingly clear that adaptive features, a hallmark of the adaptive immune system, can also be identified in the innate immune system. Such adaptations can result in the manifestation of a primed state of immune and tissue cells with a decreased activation threshold. This keeps the system poised to react quickly. Moreover, the fact that the innate immune system recognizes a wide variety of danger signals via pattern recognition receptors that often activate the same signaling pathways allows for heterologous innate immune stimulation. This implies that, for example, the innate immune response to an infection can be modified by co-infections or other innate stimuli. This "design feature" of the innate immune system has many implications for our understanding of individual susceptibility to diseases or responsiveness to therapies and vaccinations. In this article, adaptive features of the innate immune system as well as heterologous innate immunity and their implications are discussed.
Collapse
Affiliation(s)
- Stefan F Martin
- Allergy Research Group, Department of Dermatology, Medical Center - University of Freiburg, Hauptstrasse 7, 79104, Freiburg, Germany,
| |
Collapse
|
25
|
Lines JL, Sempere LF, Wang L, Pantazi E, Mak J, O’Connell S, Ceeraz S, Suriawinata AA, Yan S, Ernstoff MS, Noelle R. VISTA is an immune checkpoint molecule for human T cells. Cancer Res 2014; 74:1924-32. [PMID: 24691993 PMCID: PMC3979527 DOI: 10.1158/0008-5472.can-13-1504] [Citation(s) in RCA: 361] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
V-domain Ig suppressor of T cell activation (VISTA) is a potent negative regulator of T-cell function that is expressed on hematopoietic cells. VISTA levels are heightened within the tumor microenvironment, in which its blockade can enhance antitumor immune responses in mice. In humans, blockade of the related programmed cell death 1 (PD-1) pathway has shown great potential in clinical immunotherapy trials. Here, we report the structure of human VISTA and examine its function in lymphocyte negative regulation in cancer. VISTA is expressed predominantly within the hematopoietic compartment with highest expression within the myeloid lineage. VISTA-Ig suppressed proliferation of T cells but not B cells and blunted the production of T-cell cytokines and activation markers. Our results establish VISTA as a negative checkpoint regulator that suppresses T-cell activation, induces Foxp3 expression, and is highly expressed within the tumor microenvironment. By analogy to PD-1 and PD-L1 blockade, VISTA blockade may offer an immunotherapeutic strategy for human cancer.
Collapse
Affiliation(s)
- J. Louise Lines
- Medical Research Council Centre of Transplantation, Guy’s Hospital, King’s College London, King’s Health Partners, London, United Kingdom
- Department of Immune Regulation and Intervention, King’s College, London, SE1 9RT
| | - Lorenzo F. Sempere
- Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Li Wang
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756
| | - Eirini Pantazi
- Medical Research Council Centre of Transplantation, Guy’s Hospital, King’s College London, King’s Health Partners, London, United Kingdom
- Department of Immune Regulation and Intervention, King’s College, London, SE1 9RT
| | - Justin Mak
- Medical Research Council Centre of Transplantation, Guy’s Hospital, King’s College London, King’s Health Partners, London, United Kingdom
- Department of Immune Regulation and Intervention, King’s College, London, SE1 9RT
| | - Samuel O’Connell
- Medical Research Council Centre of Transplantation, Guy’s Hospital, King’s College London, King’s Health Partners, London, United Kingdom
- Department of Immune Regulation and Intervention, King’s College, London, SE1 9RT
| | - Sabrina Ceeraz
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756
| | | | - Shaofeng Yan
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756
| | - Marc S. Ernstoff
- Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Randolph Noelle
- Medical Research Council Centre of Transplantation, Guy’s Hospital, King’s College London, King’s Health Partners, London, United Kingdom
- Department of Immune Regulation and Intervention, King’s College, London, SE1 9RT
- Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| |
Collapse
|
26
|
Resveratrol as a natural anti-tumor necrosis factor-α molecule: implications to dendritic cells and their crosstalk with mesenchymal stromal cells. PLoS One 2014; 9:e91406. [PMID: 24614867 PMCID: PMC3948844 DOI: 10.1371/journal.pone.0091406] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 02/11/2014] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DC) are promising targets for inducing tolerance in inflammatory conditions. Thus, this study aims to investigate the effects of the natural anti-inflammatory molecule resveratrol on human DC at phenotypic and functional levels, including their capacity to recruit mesenchymal stem/stromal cells (MSC). Primary human monocyte-derived DC and bone marrow MSC were used. DC immunophenotyping revealed that small doses of resveratrol (10 µM) reduce cell activation in response to tumor necrosis factor (TNF)-α, significantly decreasing surface expression of CD83 and CD86. Functionally, IL-12/IL-23 secretion induced by TNF-α was significantly reduced by resveratrol, while IL-10 levels increased. Resveratrol also inhibited T cell proliferation, in response to TNF-α-stimulated DC. The underlying mechanism was investigated by Western blot and imaging flow cytometry (ImageStreamX), and likely involves impairment of nuclear translocation of the p65 NF-κB subunit. Importantly, results obtained demonstrate that DC are able to recruit MSC through extracellular matrix components, and that TNF-α impairs DC-mediated recruitment. Matrix metalloproteinases (MMP) produced by both cell populations were visualized by gelatin zymography. Finally, time-lapse microscopy analysis revealed a significant decrease on DC and MSC motility in co-cultures, indicating cell interaction, and TNF-α further decreased MSC motility, while resveratrol recovered it. Thus, the current study points out the potential of resveratrol as a natural anti-TNF-α drug, capable of modulating DC phenotype and function, as well as DC-mediated MSC recruitment.
Collapse
|
27
|
Highly self-reactive naive CD4 T cells are prone to differentiate into regulatory T cells. Nat Commun 2013; 4:2209. [DOI: 10.1038/ncomms3209] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 06/26/2013] [Indexed: 02/06/2023] Open
|
28
|
Kim CH. Host and microbial factors in regulation of T cells in the intestine. Front Immunol 2013; 4:141. [PMID: 23772228 PMCID: PMC3677167 DOI: 10.3389/fimmu.2013.00141] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/27/2013] [Indexed: 12/19/2022] Open
Abstract
The intestine is divided into specialized tissue areas that provide distinct microenvironments for T cells. Regulation of T-cell responses in the gut has been a major focus of recent research activities in the field. T cells in the intestine are regulated by the interplay between host and microbial factors. In the small intestine, retinoic acid (RA) is a major tissue factor that plays important roles in regulation of immune responses. In the large intestine, the influence of RA diminishes, but that of commensal bacterial products increases. RA, gut microbiota, and inflammatory mediators co-regulate differentiation, distribution, and/or effector functions of T cells. Coordinated regulation of immune responses by these factors promotes well-balanced immunity and immune tolerance. Dysregulation of this process can increase infection and inflammatory diseases.
Collapse
Affiliation(s)
- Chang H Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Center for Cancer Research, Purdue University West Lafayette, IN, USA
| |
Collapse
|
29
|
Hancock DG, Guy TV, Shklovskaya E, Fazekas de St Groth B. Experimental models to investigate the function of dendritic cell subsets: challenges and implications. Clin Exp Immunol 2013; 171:147-54. [PMID: 23286941 DOI: 10.1111/cei.12027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2012] [Indexed: 11/29/2022] Open
Abstract
The dendritic cell (DC) lineage is remarkably heterogeneous. It has been postulated that specialized DC subsets have evolved in order to select and support the multitude of possible T cell differentiation pathways. However, defining the function of individual DC subsets has proven remarkably difficult, and DC subset control of key T cell fates such as tolerance, T helper cell commitment and regulatory T cell induction is still not well understood. While the difficulty in assigning unique functions to particular DC subsets may be due to sharing of functions, it may also reflect a lack of appropriate physiological in-vivo models for studying DC function. In this paper we review the limitations associated with many of the current DC models and highlight some of the underlying difficulties involved in studying the function of murine DC subsets.
Collapse
Affiliation(s)
- D G Hancock
- Centenary Institute of Cancer Medicine and Cell Biology and the Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
30
|
Garbi N, Kreutzberg T. Dendritic cells enhance the antigen sensitivity of T cells. Front Immunol 2012; 3:389. [PMID: 23272004 PMCID: PMC3530030 DOI: 10.3389/fimmu.2012.00389] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 12/04/2012] [Indexed: 11/13/2022] Open
Abstract
Naive T cells continuously migrate between the circulatory system and lymphoid organs, where they make dynamic contacts with rare dendritic cells (DCs) that strategically form an extensive dendrite network. In such a scenario, T cells spend most of their time quickly scanning the antigenic content of multiple DCs. These interactions provide the basis for efficient adaptive responses by increasing the probability of encounters between rare antigen-specific T cells and those DCs presenting the respective cognate antigens. In the absence of foreign antigen, however, T cells show different degrees of functional sensitivity toward TCR stimulation. Scanning of MHC/self-peptide complexes by naive T cells in the absence of infection is not without consequences but it increases their subsequent response toward antigenic challenge. This indicates that TCR sensitivity in naive T cells is tuned depending on the MHC/self-peptide signals they integrate from the environment even before T cells encounter cognate antigen. DCs have emerged as key components in providing MHC/self-peptide complexes and increasing the sensitivity of T cells toward subsequent TCR triggering. In the absence of cognate antigen, DCs maintain a tonic TCR signaling and license T cells for immune synapse (IS) maturation resulting in enhanced T cell responses toward a subsequent antigen stimulation. This review discusses recent findings on this subject and highlights the importance of the DC pool size for optimal T cell awareness to foreign antigen.
Collapse
Affiliation(s)
- Natalio Garbi
- Department of Molecular Immunology, Institutes of Molecular Medicine and Experimental Immunology, University of Bonn Bonn, Germany
| | | |
Collapse
|
31
|
Lynch JN, Donermeyer DL, Weber KS, Kranz DM, Allen PM. Subtle changes in TCRα CDR1 profoundly increase the sensitivity of CD4 T cells. Mol Immunol 2012; 53:283-94. [PMID: 22982754 DOI: 10.1016/j.molimm.2012.08.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 08/15/2012] [Accepted: 08/18/2012] [Indexed: 10/27/2022]
Abstract
Changes in the peptide and MHC molecules have been extensively examined for how they alter T cell activation, but many fewer studies have examined the TCR. Structural studies of how TCR differences alter T cell specificity have focused on broad variation in the CDR3 loops. However, changes in the CDR1 and 2 loops can also alter TCR recognition of pMHC. In this study we focus on two mutations in the CDR1α loop of the TCR that increased the affinity of a TCR for agonist Hb(64-76)/I-E(k) by increasing the on-rate of the reaction. These same mutations also conferred broader recognition of altered peptide ligands. TCR transgenic mice expressing the CDR1α mutations had altered thymic selection, as most of the T cells were negatively selected compared to T cells expressing the wildtype TCR. The few T cells that escaped negative selection and were found in the periphery were rendered anergic, thereby avoiding autoimmunity. T cells with the CDR1α mutations were completely deleted in the presence of Hb(64-76) as an endogenous peptide. Interestingly, the wildtype T cells were not eliminated, identifying a threshold affinity for negative selection where a 3-fold increase in affinity is the difference between incomplete and complete deletion. Overall, these studies highlight how small changes in the TCR can increase the affinity of TCR:pMHC but with the consequences of skewing selection and producing an unresponsive T cell.
Collapse
Affiliation(s)
- Jennifer N Lynch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | | | | | | | | |
Collapse
|
32
|
Lewis KL, Reizis B. Dendritic cells: arbiters of immunity and immunological tolerance. Cold Spring Harb Perspect Biol 2012; 4:a007401. [PMID: 22855722 DOI: 10.1101/cshperspect.a007401] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) link innate immune sensing of the environment to the initiation of adaptive immune responses. Given their supreme capacity to interact with and present antigen to T cells, DCs have been proposed as key mediators of immunological tolerance in the steady state. However, recent evidence suggests that the role of DCs in central and peripheral T-cell tolerance is neither obligate nor dominant. Instead, DCs appear to regulate multiple aspects of T-cell physiology including tonic antigen receptor signaling, priming of effector T-cell response, and the maintenance of regulatory T cells. These diverse contributions of DCs may reflect the significant heterogeneity and "division of labor" observed between and within distinct DC subsets. The emerging complex role of different DC subsets should form the conceptual basis of DC-based therapeutic approaches toward induction of tolerance or immunization.
Collapse
Affiliation(s)
- Kanako L Lewis
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, 10032, USA
| | | |
Collapse
|
33
|
Release of dendritic cells from cognate CD4+ T-cell recognition results in impaired peripheral tolerance and fatal cytotoxic T-cell mediated autoimmunity. Proc Natl Acad Sci U S A 2012; 109:9059-64. [PMID: 22615402 DOI: 10.1073/pnas.1110620109] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Resting dendritic cells (DCs) induce tolerance of peripheral T cells that have escaped thymic negative selection and thus contribute significantly to protection against autoimmunity. We recently showed that CD4(+)Foxp3(+) regulatory T cells (Tregs) are important for maintaining the steady-state phenotype of DCs and their tolerizing capacity in vivo. We now provide evidence that DC activation in the absence of Tregs is a direct consequence of missing DC-Treg interactions rather than being secondary to generalized autoimmunity in Treg-less mice. We show that DCs that lack MHC class II and thus cannot make cognate interactions with CD4(+) T cells are completely unable to induce peripheral CD8(+) T-cell tolerance. Consequently, mice in which interactions between DC and CD4(+) T cells are not possible develop spontaneous and fatal cytotoxic T lymphocyte-mediated autoimmunity.
Collapse
|
34
|
Delpoux A, Poitrasson-Rivière M, Le Campion A, Pommier A, Yakonowsky P, Jacques S, Letourneur F, Randriamampita C, Lucas B, Auffray C. Foxp3-independent loss of regulatory CD4+T-cell suppressive capacities induced by self-deprivation. Eur J Immunol 2012; 42:1237-49. [DOI: 10.1002/eji.201142148] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Arnaud Delpoux
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Maud Poitrasson-Rivière
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Armelle Le Campion
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Arnaud Pommier
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Philippe Yakonowsky
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Sébastien Jacques
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Franck Letourneur
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Clotilde Randriamampita
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Bruno Lucas
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| | - Cédric Auffray
- CNRS UMR8104; Cochin Hospital; Paris France
- INSERM U567; Cochin Hospital; Paris France
- Paris Descartes University; Cochin Hospital; Paris France
| |
Collapse
|
35
|
Allen CT, Judd NP, Bui JD, Uppaluri R. The clinical implications of antitumor immunity in head and neck cancer. Laryngoscope 2011; 122:144-57. [DOI: 10.1002/lary.21913] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
36
|
Normal T cell homeostasis: the conversion of naive cells into memory-phenotype cells. Nat Immunol 2011; 12:478-84. [PMID: 21739670 DOI: 10.1038/ni.2018] [Citation(s) in RCA: 359] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Weak T cell antigen receptor (TCR) signals from contact with self ligands act in synergy with antiapoptotic signals induced by interleukin 7 (IL-7) to promote the survival of naive T cells in a resting state. The amount of background TCR signaling in naive T cells is set by post-thymic TCR tuning and operates at an intensity just below that required to induce entry into the cell cycle. Costimulation from higher concentrations of IL-7 and other common γ-chain cytokines can induce T cells to undergo homeostatic proliferation and conversion into cells with a memory phenotype; many of these memory phenotype cells may be the progeny of cells responding to self antigens. The molecular mechanisms that control the conversion of naive resting T cells into memory-phenotype cells TCR-dependent in normal animals are beginning to be understood.
Collapse
|
37
|
Garbi N, Hämmerling GJ. Co-stimulation by dendritic cells maintains the peripheral pool of Tregs. Eur J Immunol 2011; 41:282-5. [DOI: 10.1002/eji.201041335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
38
|
The stromal and haematopoietic antigen-presenting cells that reside in secondary lymphoid organs. Nat Rev Immunol 2010; 10:813-25. [DOI: 10.1038/nri2886] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|