1
|
Lo WL, Kuhlmann M, Rizzuto G, Ekiz HA, Kolawole EM, Revelo MP, Andargachew R, Li Z, Tsai YL, Marson A, Evavold BD, Zehn D, Weiss A. A single-amino acid substitution in the adaptor LAT accelerates TCR proofreading kinetics and alters T-cell selection, maintenance and function. Nat Immunol 2023; 24:676-689. [PMID: 36914891 PMCID: PMC10063449 DOI: 10.1038/s41590-023-01444-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/25/2023] [Indexed: 03/14/2023]
Abstract
Mature T cells must discriminate between brief interactions with self-peptides and prolonged binding to agonists. The kinetic proofreading model posits that certain T-cell antigen receptor signaling nodes serve as molecular timers to facilitate such discrimination. However, the physiological significance of this regulatory mechanism and the pathological consequences of disrupting it are unknown. Here we report that accelerating the normally slow phosphorylation of the linker for activation of T cells (LAT) residue Y136 by introducing an adjacent Gly135Asp alteration (LATG135D) disrupts ligand discrimination in vivo. The enhanced self-reactivity of LATG135D T cells triggers excessive thymic negative selection and promotes T-cell anergy. During Listeria infection, LATG135D T cells expand more than wild-type counterparts in response to very weak stimuli but display an imbalance between effector and memory responses. Moreover, despite their enhanced engagement of central and peripheral tolerance mechanisms, mice bearing LATG135D show features associated with autoimmunity and immunopathology. Our data reveal the importance of kinetic proofreading in balancing tolerance and immunity.
Collapse
Affiliation(s)
- Wan-Lin Lo
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA.
| | - Miriam Kuhlmann
- Division of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Gabrielle Rizzuto
- Human Oncology and Pathogenesis Program, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - H Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce, Turkey
| | - Elizabeth M Kolawole
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Monica P Revelo
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Rakieb Andargachew
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Zhongmei Li
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Yuan-Li Tsai
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Brian D Evavold
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany.
| | - Arthur Weiss
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Vidard L. 4-1BB and cytokines trigger human NK, γδ T, and CD8 + T cell proliferation and activation, but are not required for their effector functions. Immun Inflamm Dis 2023; 11:e749. [PMID: 36705415 PMCID: PMC9753824 DOI: 10.1002/iid3.749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION This study was designed to compare the costimulatory molecules and cytokines required to trigger the proliferation and activation of natural killer (NK), γδ T, and CD8+ T cells, and gain in-depth insight into the mechanisms shifting tolerance to immunity. METHODS K562-derived artificial antigen-presenting cells (aAPCs); that is, K562 forced to express CD86 and 4-1BBL costimulatory receptors, in the presence of cytokines, were used to mimic dendritic cells (DCs) and provide signals to support the proliferation and activation of NK, γδ T, and CD8+ T cells. RESULTS Three signals are required to trigger optimal proliferation in MART-1-specific CD8+ T cells: activation of T-cell receptors (TCRs) by the major histocompatibility complex (MHC) I/peptide complexes (signal 1); 4-1BB engagement (signal 2); and IL-15 and IL-21 receptor co-signaling (signal 3). NK and γδ T cell proliferation also require three signals, but the precise nature of signal 1 involving cell-to-cell contact was not determined. Once they become effectors, only signal 1 determines the sensitivity or resistance of the target cells to cytolysis by killer lymphocytes. When freshly purified, none had effector functions, except the NK cells, which could be activated by CD16 engagement. CONCLUSIONS Therefore, lymphocytes committed to kill are produced as inactive precursors, and the license to kill is delivered by three signals, allowing for extensive proliferation and effector function acquisition. This data challenges the paradigm of anergy and supports the danger signal theory originally proposed by Polly Matzinger, which states that killer cells are tolerant by default, thereby protecting the mammalian body from autoimmunity.
Collapse
Affiliation(s)
- Laurent Vidard
- Department of Immuno‐OncologySanofiVitry‐sur‐SeineFrance
| |
Collapse
|
3
|
Hodgson R, Xu X, Anzilotti C, Deobagkar-Lele M, Crockford TL, Kepple JD, Cawthorne E, Bhandari A, Cebrian-Serrano A, Wilcock MJ, Davies B, Cornall RJ, Bull KR. NDRG1 is induced by antigen-receptor signaling but dispensable for B and T cell self-tolerance. Commun Biol 2022; 5:1216. [PMID: 36357486 PMCID: PMC9649591 DOI: 10.1038/s42003-022-04118-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Peripheral tolerance prevents the initiation of damaging immune responses by autoreactive lymphocytes. While tolerogenic mechanisms are tightly regulated by antigen-dependent and independent signals, downstream pathways are incompletely understood. N-myc downstream-regulated gene 1 (NDRG1), an anti-cancer therapeutic target, has previously been implicated as a CD4+ T cell clonal anergy factor. By RNA-sequencing, we identified Ndrg1 as the third most upregulated gene in anergic, compared to naïve follicular, B cells. Ndrg1 is upregulated by B cell receptor activation (signal one) and suppressed by co-stimulation (signal two), suggesting that NDRG1 may be important in B cell tolerance. However, though Ndrg1-/- mice have a neurological defect mimicking NDRG1-associated Charcot-Marie-Tooth (CMT4d) disease, primary and secondary immune responses were normal. We find that B cell tolerance is maintained, and NDRG1 does not play a role in downstream responses during re-stimulation of in vivo antigen-experienced CD4+ T cells, demonstrating that NDGR1 is functionally redundant for lymphocyte anergy.
Collapse
Affiliation(s)
- Rose Hodgson
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Xijin Xu
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Consuelo Anzilotti
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mukta Deobagkar-Lele
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tanya L Crockford
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jessica D Kepple
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Eleanor Cawthorne
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Aneesha Bhandari
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alberto Cebrian-Serrano
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Martin J Wilcock
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Benjamin Davies
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Richard J Cornall
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Katherine R Bull
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Korman AJ, Garrett-Thomson SC, Lonberg N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat Rev Drug Discov 2021; 21:509-528. [PMID: 34937915 DOI: 10.1038/s41573-021-00345-8] [Citation(s) in RCA: 302] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 12/11/2022]
Abstract
Cancer immunity, and the potential for cancer immunotherapy, have been topics of scientific discussion and experimentation for over a hundred years. Several successful cancer immunotherapies - such as IL-2 and interferon-α (IFNα) - have appeared over the past 30 years. However, it is only in the past decade that immunotherapy has made a broad impact on patient survival in multiple high-incidence cancer indications. The emergence of immunotherapy as a new pillar of cancer treatment (adding to surgery, radiation, chemotherapy and targeted therapies) is due to the success of immune checkpoint blockade (ICB) drugs, the first of which - ipilimumab - was approved in 2011. ICB drugs block receptors and ligands involved in pathways that attenuate T cell activation - such as cytotoxic T lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD1) and its ligand, PDL1 - and prevent, or reverse, acquired peripheral tolerance to tumour antigens. In this Review we mark the tenth anniversary of the approval of ipilimumab and discuss the foundational scientific history of ICB, together with the history of the discovery, development and elucidation of the mechanism of action of the first generation of drugs targeting the CTLA4 and PD1 pathways.
Collapse
|
5
|
Co DO, Hogan LH, Karman J, Herbath M, Fabry Z, Sandor M. T Cell Interactions in Mycobacterial Granulomas: Non-Specific T Cells Regulate Mycobacteria-Specific T Cells in Granulomatous Lesions. Cells 2021; 10:cells10123285. [PMID: 34943793 PMCID: PMC8699651 DOI: 10.3390/cells10123285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/16/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022] Open
Abstract
Infections with pathogenic mycobacteria are controlled by the formation of a unique structure known as a granuloma. The granuloma represents a host–pathogen interface where bacteria are killed and confined by the host response, but also where bacteria persist. Previous work has demonstrated that the T cell repertoire is heterogenous even at the single granuloma level. However, further work using pigeon cytochrome C (PCC) epitope-tagged BCG (PCC-BCG) and PCC-specific 5CC7 RAG−/− TCR transgenic (Tg) mice has demonstrated that a monoclonal T cell population is able to control infection. At the chronic stage of infection, granuloma-infiltrating T cells remain highly activated in wild-type mice, while T cells in the monoclonal T cell mice are anergic. We hypothesized that addition of an acutely activated non-specific T cell to the monoclonal T cell system could recapitulate the wild-type phenotype. Here we report that activated non-specific T cells have access to the granuloma and deliver a set of cytokines and chemokines to the lesions. Strikingly, non-specific T cells rescue BCG-specific T cells from anergy and enhance the function of BCG-specific T cells in the granuloma in the chronic phase of infection when bacterial antigen load is low. In addition, we find that these same non-specific T cells have an inhibitory effect on systemic BCG-specific T cells. Taken together, these data suggest that T cells non-specific for granuloma-inducing agents can alter the function of granuloma-specific T cells and have important roles in mycobacterial immunity and other granulomatous disorders.
Collapse
Affiliation(s)
- Dominic O. Co
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Laura H. Hogan
- The Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Jozsef Karman
- Cambridge Research Center, Abbvie, Inc., Cambridge, MA 02139, USA;
| | - Melinda Herbath
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.H.); (Z.F.)
| | - Zsuzsanna Fabry
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.H.); (Z.F.)
| | - Matyas Sandor
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.H.); (Z.F.)
- Correspondence: ; Tel.: +1-(608)-265-8715
| |
Collapse
|
6
|
Maulloo CD, Cao S, Watkins EA, Raczy MM, Solanki AS, Nguyen M, Reda JW, Shim HN, Wilson DS, Swartz MA, Hubbell JA. Lymph Node-Targeted Synthetically Glycosylated Antigen Leads to Antigen-Specific Immunological Tolerance. Front Immunol 2021; 12:714842. [PMID: 34630389 PMCID: PMC8498032 DOI: 10.3389/fimmu.2021.714842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Inverse vaccines that tolerogenically target antigens to antigen-presenting cells (APCs) offer promise in prevention of immunity to allergens and protein drugs and treatment of autoimmunity. We have previously shown that targeting hepatic APCs through intravenous injection of synthetically glycosylated antigen leads to effective induction of antigen-specific immunological tolerance. Here, we demonstrate that targeting these glycoconjugates to lymph node (LN) APCs under homeostatic conditions leads to local and increased accumulation in the LNs compared to unmodified antigen and induces a tolerogenic state both locally and systemically. Subcutaneous administration directs the polymeric glycoconjugate to the draining LN, where the glycoconjugated antigen generates robust antigen-specific CD4+ and CD8+ T cell tolerance and hypo-responsiveness to antigenic challenge via a number of mechanisms, including clonal deletion, anergy of activated T cells, and expansion of regulatory T cells. Lag-3 up-regulation on CD4+ and CD8+ T cells represents an essential mechanism of suppression. Additionally, presentation of antigen released from the glycoconjugate to naïve T cells is mediated mainly by LN-resident CD8+ and CD11b+ dendritic cells. Thus, here we demonstrate that antigen targeting via synthetic glycosylation to impart affinity for APC scavenger receptors generates tolerance when LN dendritic cells are the cellular target.
Collapse
Affiliation(s)
- Chitavi D. Maulloo
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Elyse A. Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Michal M. Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Ani. S. Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, United States
| | - Mindy Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Joseph W. Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - D. Scott Wilson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, United States
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Committee on Immunology, University of Chicago, Chicago, IL, United States
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Committee on Immunology, University of Chicago, Chicago, IL, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States
| |
Collapse
|
7
|
Exploring the Pathogenic Role and Therapeutic Implications of Interleukin 2 in Autoimmune Hepatitis. Dig Dis Sci 2021; 66:2493-2512. [PMID: 32833154 DOI: 10.1007/s10620-020-06562-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022]
Abstract
Interleukin 2 is essential for the expansion of regulatory T cells, and low-dose recombinant interleukin 2 has improved the clinical manifestations of diverse autoimmune diseases in preliminary studies. The goals of this review are to describe the actions of interleukin 2 and its receptor, present preliminary experiences with low-dose interleukin 2 in the treatment of diverse autoimmune diseases, and evaluate its potential as a therapeutic intervention in autoimmune hepatitis. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Interleukin 2 is critical for the thymic selection, peripheral expansion, induction, and survival of regulatory T cells, and it is also a growth factor for activated T cells and natural killer cells. Interleukin 2 activates the signal transducer and activator of transcription 5 after binding with its trimeric receptor on regulatory T cells. Immune suppressor activity is increased; anti-inflammatory interleukin 10 is released; pro-inflammatory interferon-gamma is inhibited; and activation-induced apoptosis of CD8+ T cells is upregulated. Preliminary experiences with cyclic injections of low-dose recombinant interleukin 2 in diverse autoimmune diseases have demonstrated increased numbers of circulating regulatory T cells, preserved regulatory function, improved clinical manifestations, and excellent tolerance. Similar improvements have been recognized in one of two patients with refractory autoimmune hepatitis. In conclusion, interferon 2 has biological actions that favor the immune suppressor functions of regulatory T cells, and low-dose regimens in preliminary studies encourage its rigorous investigation in autoimmune hepatitis.
Collapse
|
8
|
Sim JH, Kim JH, Park AK, Lee J, Kim KM, Shin HM, Kim M, Choi K, Choi EY, Kang I, Lee DS, Kim HR. IL-7Rα low CD8 + T Cells from Healthy Individuals Are Anergic with Defective Glycolysis. THE JOURNAL OF IMMUNOLOGY 2020; 205:2968-2978. [PMID: 33106337 DOI: 10.4049/jimmunol.1901470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 09/22/2020] [Indexed: 11/19/2022]
Abstract
Effector memory (EM) CD8+ T cells expressing lower levels of IL-7R α (IL-7Rαlow) from healthy individuals are partly compromised in vitro, but the identity of these cells has remained unclear. In this study, we demonstrate that human IL-7Rαlow EM CD8+ T cells are naturally occurring anergic cells in vivo and impaired in proliferation and IL-2 production but competent in IFN-γ and TNF-α production, a state that can be restored by IL-2 stimulation. IL-7Rαlow EM CD8+ T cells show decreased expression of GATA3 and c-MYC and are defective in metabolic reprogramming toward glycolysis, a process required for the proliferation of T cells. However, IL-7Rαlow EM CD8+ T cells can proliferate with TCR stimulation in the presence of IL-2 and IL-15, suggesting that these cells can be restored to normality or increased activity by inflammatory conditions and may serve as a reservoir for functional immunity.
Collapse
Affiliation(s)
- Ji Hyun Sim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jin-Hee Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28150, Chungbuk, Republic of Korea
| | - Ae Kyung Park
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, College of Pharmacy, Sunchon National University, Suncheon 57922, Jeonnam, Republic of Korea
| | - Jeeyun Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Hyun Mu Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Wide River Institute of Immunology, Seoul National University, Hongcheon 25159, Republic of Korea
| | - Minji Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Kyungho Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; and
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Insoo Kang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Dong-Sup Lee
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Wide River Institute of Immunology, Seoul National University, Hongcheon 25159, Republic of Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hang-Rae Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; .,Medical Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Wide River Institute of Immunology, Seoul National University, Hongcheon 25159, Republic of Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
9
|
Rapid in vitro generation of bona fide exhausted CD8+ T cells is accompanied by Tcf7 promotor methylation. PLoS Pathog 2020; 16:e1008555. [PMID: 32579593 PMCID: PMC7340326 DOI: 10.1371/journal.ppat.1008555] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 07/07/2020] [Accepted: 04/15/2020] [Indexed: 12/23/2022] Open
Abstract
Exhaustion is a dysfunctional state of cytotoxic CD8+ T cells (CTL) observed in chronic infection and cancer. Current in vivo models of CTL exhaustion using chronic viral infections or cancer yield very few exhausted CTL, limiting the analysis that can be done on these cells. Establishing an in vitro system that rapidly induces CTL exhaustion would therefore greatly facilitate the study of this phenotype, identify the truly exhaustion-associated changes and allow the testing of novel approaches to reverse or prevent exhaustion. Here we show that repeat stimulation of purified TCR transgenic OT-I CTL with their specific peptide induces all the functional (reduced cytokine production and polyfunctionality, decreased in vivo expansion capacity) and phenotypic (increased inhibitory receptors expression and transcription factor changes) characteristics of exhaustion. Importantly, in vitro exhausted cells shared the transcriptomic characteristics of the gold standard of exhaustion, CTL from LCMV cl13 infections. Gene expression of both in vitro and in vivo exhausted CTL was distinct from T cell anergy. Using this system, we show that Tcf7 promoter DNA methylation contributes to TCF1 downregulation in exhausted CTL. Thus this novel in vitro system can be used to identify genes and signaling pathways involved in exhaustion and will facilitate the screening of reagents that prevent/reverse CTL exhaustion.
Collapse
|
10
|
Itoh A, Ortiz L, Kachapati K, Wu Y, Adams D, Bednar K, Mukherjee S, Chougnet C, Mittler RS, Chen YG, Dolan L, Ridgway WM. Soluble CD137 Ameliorates Acute Type 1 Diabetes by Inducing T Cell Anergy. Front Immunol 2019; 10:2566. [PMID: 31787971 PMCID: PMC6853870 DOI: 10.3389/fimmu.2019.02566] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/16/2019] [Indexed: 12/21/2022] Open
Abstract
We show here that soluble CD137 (sCD137), the alternately spliced gene product of Tnfsfr9, effectively treats acute type 1 diabetes (T1D) in nonobese diabetic (NOD) mice. sCD137 significantly delayed development of end-stage disease, preserved insulin+ islet beta cells, and prevented progression to end-stage T1D in some mice. We demonstrate that sCD137 induces CD4+ T cell anergy, suppressing antigen-specific T cell proliferation and IL-2/IFN-γ secretion. Exogenous IL-2 reversed the sCD137 anergy effect. sCD137 greatly reduces inflammatory cytokine production by CD8 effector memory T cells, critical mediators of beta cell damage. We demonstrate that human T1D patients have decreased serum sCD137 compared to age-matched controls (as do NOD mice compared to NOD congenic mice expressing a protective Tnfsfr9 allele), that human sCD137 is secreted by regulatory T cells (Tregs; as in mice), and that human sCD137 induces T cell suppression in human T cells. These findings provide a rationale for further investigation of sCD137 as a treatment for T1D and other T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Arata Itoh
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Lorenzo Ortiz
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Kritika Kachapati
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Yuehong Wu
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David Adams
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Kyle Bednar
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Claire Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Robert S Mittler
- Department of Surgery, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Atlanta, GA, United States
| | - Yi-Guang Chen
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Laurence Dolan
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - William M Ridgway
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
11
|
Jayaraman S, Prabhakar BS. Immune Tolerance in Autoimmune Central Nervous System Disorders. CONTEMPORARY CLINICAL NEUROSCIENCE 2019. [PMCID: PMC7121051 DOI: 10.1007/978-3-030-19515-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
12
|
Woroniecka KI, Rhodin KE, Chongsathidkiet P, Keith KA, Fecci PE. T-cell Dysfunction in Glioblastoma: Applying a New Framework. Clin Cancer Res 2018; 24:3792-3802. [PMID: 29593027 DOI: 10.1158/1078-0432.ccr-18-0047] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/01/2018] [Accepted: 03/26/2018] [Indexed: 02/06/2023]
Abstract
A functional, replete T-cell repertoire is an integral component to adequate immune surveillance and to the initiation and maintenance of productive antitumor immune responses. Glioblastoma (GBM), however, is particularly adept at sabotaging antitumor immunity, eliciting severe T-cell dysfunction that is both qualitative and quantitative. Understanding and countering such dysfunction are among the keys to harnessing the otherwise stark potential of anticancer immune-based therapies. Although T-cell dysfunction in GBM has been long described, newer immunologic frameworks now exist for reclassifying T-cell deficits in a manner that better permits their study and reversal. Herein, we divide and discuss the various T-cell deficits elicited by GBM within the context of the five relevant categories: senescence, tolerance, anergy, exhaustion, and ignorance. Categorization is appropriately made according to the molecular bases of dysfunction. Likewise, we review the mechanisms by which GBM elicits each mode of T-cell dysfunction and discuss the emerging immunotherapeutic strategies designed to overcome them. Clin Cancer Res; 24(16); 3792-802. ©2018 AACR.
Collapse
Affiliation(s)
- Karolina I Woroniecka
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Kristen E Rhodin
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Pakawat Chongsathidkiet
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Kristin A Keith
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Peter E Fecci
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina. .,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
13
|
Woodham AW, Yan L, Skeate JG, van der Veen D, Brand HH, Wong MK, Da Silva DM, Kast WM. T cell ignorance is bliss: T cells are not tolerized by Langerhans cells presenting human papillomavirus antigens in the absence of costimulation. PAPILLOMAVIRUS RESEARCH 2018; 2:21-30. [PMID: 27182559 PMCID: PMC4862606 DOI: 10.1016/j.pvr.2016.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human papillomavirus type 16 (HPV16) infections are intra-epithelial, and thus, HPV16 is known to interact with Langerhans cells (LCs), the resident epithelial antigen-presenting cells (APCs). The current paradigm for APC-mediated induction of T cell anergy is through delivery of T cell receptor signals via peptides on MHC molecules (signal 1), but without costimulation (signal 2). We previously demonstrated that LCs exposed to HPV16 in vitro present HPV antigens to T cells without costimulation, but it remained uncertain if such T cells would remain ignorant, become anergic, or in the case of CD4+ T cells, differentiate into Tregs. Here we demonstrate that Tregs were not induced by LCs presenting only signal 1, and through a series of in vitro immunizations show that CD8+ T cells receiving signal 1+2 from LCs weeks after consistently receiving signal 1 are capable of robust effector functions. Importantly, this indicates that T cells are not tolerized but instead remain ignorant to HPV, and are activated given the proper signals.
Collapse
Affiliation(s)
- Andrew W Woodham
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, California, United States of America
| | - Lisa Yan
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, California, United States of America
| | - Joseph G Skeate
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, California, United States of America
| | | | - Heike H Brand
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Michael K Wong
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Diane M Da Silva
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America; Department of Obstetrics & Gynecology, University of Southern California, Los Angeles, California, United States of America
| | - W Martin Kast
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, California, United States of America; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America; Department of Obstetrics & Gynecology, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
14
|
Dillinger B, Ahmadi-Erber S, Soukup K, Halfmann A, Schrom S, Vanhove B, Steinberger P, Geyeregger R, Ladisch S, Dohnal AM. CD28 Blockade Ex Vivo Induces Alloantigen-Specific Immune Tolerance but Preserves T-Cell Pathogen Reactivity. Front Immunol 2017; 8:1152. [PMID: 28979262 PMCID: PMC5611377 DOI: 10.3389/fimmu.2017.01152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022] Open
Abstract
Donor T-cells contribute to reconstitution of protective immunity after allogeneic hematopoietic stem cell transplantation (HSCT) but must acquire specific tolerance against recipient alloantigens to avoid life-threatening graft-versus-host disease (GvHD). Systemic immunosuppressive drugs may abrogate severe GvHD, but this also impedes memory responses to invading pathogens. Here, we tested whether ex vivo blockade of CD28 co-stimulation can enable selective T-cell tolerization to alloantigens by facilitating CD80/86-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling. Treatment of human allogeneic dendritic cell/T-cell co-cultures with a human CD28 blocking antibody fragment (α-huCD28) significantly abrogated subsequent allospecific immune responses, seen by decreased T-cell proliferation and of type 1 cytokine (IFN-γ and IL-2) expression. Allo-tolerization persisted after discontinuation of CD28 blockade and secondary alloantigen stimulation, as confirmed by enhanced CTLA-4 and PD-1 immune checkpoint signaling. However, T-cells retained reactivity to pathogens, supported by clonotyping of neo-primed and cross-reactive T-cells specific for Candida albicans or third-party antigens using deep sequencing analysis. In an MHC-mismatched murine model, we tolerized C57BL/6 T-cells by ex vivo exposure to a murine single chain Fv specific for CD28 (α-muCD28). Infusion of these cells, after α-muCD28 washout, into bone marrow-transplanted BALB/c mice caused allo-tolerance and did not induce GvHD-associated hepatic pathology. We conclude that selective CD28 blockade ex vivo can allow the generation of stably allo-tolerized T-cells that in turn do not induce graft-versus-host reactions while maintaining pathogen reactivity. Hence, CD28 co-stimulation blockade of donor T-cells may be a useful therapeutic approach to support the immune system after HSCT.
Collapse
Affiliation(s)
- Barbara Dillinger
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Sarah Ahmadi-Erber
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Klara Soukup
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Angela Halfmann
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Silke Schrom
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Bernard Vanhove
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Rene Geyeregger
- Clinical Cell Biology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Stephan Ladisch
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center, Washington, DC, United States
| | - Alexander Michael Dohnal
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| |
Collapse
|
15
|
Sim JH, Kim KS, Park H, Kim KJ, Lin H, Kim TJ, Shin HM, Kim G, Lee DS, Park CW, Lee DH, Kang I, Kim SJ, Cho CH, Doh J, Kim HR. Differentially Expressed Potassium Channels Are Associated with Function of Human Effector Memory CD8 + T Cells. Front Immunol 2017; 8:859. [PMID: 28791017 PMCID: PMC5522836 DOI: 10.3389/fimmu.2017.00859] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 07/07/2017] [Indexed: 12/12/2022] Open
Abstract
The voltage-gated potassium channel, Kv1.3, and the Ca2+-activated potassium channel, KCa3.1, regulate membrane potentials in T cells, thereby controlling T cell activation and cytokine production. However, little is known about the expression and function of potassium channels in human effector memory (EM) CD8+ T cells that can be further divided into functionally distinct subsets based on the expression of the interleukin (IL)-7 receptor alpha (IL-7Rα) chain. Herein, we investigated the functional expression and roles of Kv1.3 and KCa3.1 in EM CD8+ T cells that express high or low levels of the IL-7 receptor alpha chain (IL-7Rαhigh and IL-7Rαlow, respectively). In contrast to the significant activity of Kv1.3 and KCa3.1 in IL-7Rαhigh EM CD8+ T cells, IL-7Rαlow EM CD8+ T cells showed lower expression of Kv1.3 and insignificant expression of KCa3.1. Kv1.3 was involved in the modulation of cell proliferation and IL-2 production, whereas KCa3.1 affected the motility of EM CD8+ T cells. The lower motility of IL-7Rαlow EM CD8+ T cells was demonstrated using transendothelial migration and motility assays with intercellular adhesion molecule 1- and/or chemokine stromal cell-derived factor-1α-coated surfaces. Consistent with the lower migration property, IL-7Rαlow EM CD8+ T cells were found less frequently in human skin. Stimulating IL-7Rαlow EM CD8+ T cells with IL-2 or IL-15 increased their motility and recovery of KCa3.1 activity. Our findings demonstrate that Kv1.3 and KCa3.1 are differentially involved in the functions of EM CD8+ T cells. The weak expression of potassium channels in IL-7Rαlow EM CD8+ T cells can be revived by stimulation with IL-2 or IL-15, which restores the associated functions. This study suggests that IL-7Rαhigh EM CD8+ T cells with functional potassium channels may serve as a reservoir for effector CD8+ T cells during peripheral inflammation.
Collapse
Affiliation(s)
- Ji Hyun Sim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyung Soo Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyoungjun Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Kyung-Jin Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea
| | - Haiyue Lin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae-Joo Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun Mu Shin
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Gwanghun Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong-Sup Lee
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Chan-Wook Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, South Korea
| | - Insoo Kang
- Department of Internal Medicine, Section of Rheumatology, Yale University School of Medicine, New Haven, CT, United States
| | - Sung Joon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Chung-Hyun Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Junsang Doh
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Hang-Rae Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
16
|
Dáňová K, Grohová A, Strnadová P, Funda DP, Šumník Z, Lebl J, Cinek O, Průhová Š, Koloušková S, Obermannová B, Petruželková L, Šedivá A, Fundová P, Buschard K, Špíšek R, Palová-Jelínková L. Tolerogenic Dendritic Cells from Poorly Compensated Type 1 Diabetes Patients Have Decreased Ability To Induce Stable Antigen-Specific T Cell Hyporesponsiveness and Generation of Suppressive Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2016; 198:729-740. [PMID: 27927966 DOI: 10.4049/jimmunol.1600676] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/29/2016] [Indexed: 12/15/2022]
Abstract
Tolerogenic dendritic cells (tolDCs) may offer an interesting intervention strategy to re-establish Ag-specific tolerance in autoimmune diseases, including type 1 diabetes (T1D). T1D results from selective destruction of insulin-producing β cells leading to hyperglycemia that, in turn, specifically affects a patient's immune system. In this study, we prepared monocyte-derived tolDCs modulated by dexamethasone and vitamin D2 from 31 T1D patients with optimal glycemic control and 60 T1D patients with suboptimal glycemic control and assessed their tolerogenic properties in correlation with metabolic state of patients. tolDCs differentiated from both groups of patients acquired a regulatory phenotype and an anti-inflammatory profile. Interestingly, tolDCs from well-controlled patients expressed higher levels of inhibitory molecules IL-T3 and PD-L1. Additionally, glutamic acid decarboxylase (GAD)65-loaded tolDCs from well-controlled patients decreased significantly primary Th1/Th17 responses, induced stable GAD65-specific T cell hyporesponsiveness, and suppressed markedly control DC-induced GAD65-specific T cell activation compared with poorly controlled patients. The ability of tolDCs from poorly controlled patients to induce durable GAD65-specific T cell hyporesponsiveness was reversed once the control of glycemia improved. In both groups of patients, tolDCs were able to induce regulatory T cells from autologous naive CD4+ T cells. However, regulatory T cells from well-controlled patients had better suppressive abilities. The functionality of tolDCs was confirmed in the adoptive transfer model of NOD-SCID mice where tolDCs delayed diabetes onset. These results suggest that metabolic control of T1D affects the functional characteristics of tolDCs and subsequent effector T cell responses. Metabolic control may be relevant for refining inclusion criteria of clinical trials in the settings of T1D.
Collapse
Affiliation(s)
- Klára Dáňová
- Sotio a.s., 170 00 Prague, Czech Republic.,Department of Immunology, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic
| | - Anna Grohová
- Sotio a.s., 170 00 Prague, Czech Republic.,Department of Immunology, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic
| | | | - David P Funda
- Department of Immunology and Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, v.v.i., 142 20 Prague, Czech Republic
| | - Zdeněk Šumník
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic; and
| | - Jan Lebl
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic; and
| | - Ondřej Cinek
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic; and
| | - Štěpánka Průhová
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic; and
| | - Stanislava Koloušková
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic; and
| | - Barbora Obermannová
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic; and
| | - Lenka Petruželková
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic; and
| | - Anna Šedivá
- Department of Immunology, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic
| | - Petra Fundová
- Department of Immunology and Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, v.v.i., 142 20 Prague, Czech Republic
| | - Karsten Buschard
- The Bartholin Institute, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Radek Špíšek
- Sotio a.s., 170 00 Prague, Czech Republic.,Department of Immunology, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic
| | - Lenka Palová-Jelínková
- Sotio a.s., 170 00 Prague, Czech Republic; .,Department of Immunology, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 150 06 Prague, Czech Republic
| |
Collapse
|
17
|
Ohtsuka S, Ogawa S, Wakamatsu E, Abe R. Cell cycle arrest caused by MEK/ERK signaling is a mechanism for suppressing growth of antigen-hyperstimulated effector T cells. Int Immunol 2016; 28:547-557. [PMID: 27543653 DOI: 10.1093/intimm/dxw037] [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: 06/29/2016] [Accepted: 08/17/2016] [Indexed: 12/17/2022] Open
Abstract
Suppression of T-cell growth is an important mechanism for establishment of self-tolerance and prevention of unwanted prolonged immune responses that may cause tissue damage. Although negative selection of potentially self-reactive T cells in the thymus as well as in peripheral tissues has been extensively investigated and well documented, regulatory mechanisms to dampen proliferation of antigen-specific effector T cells in response to antigen stimulation remain largely unknown. Thus, in this work, we focus on the identification of growth suppression mechanisms of antigen-specific effector T cells. In order to address this issue, we investigated the cellular and molecular events in growth suppression of an ovalbumin (OVA)-specific T-cell clone after stimulation with a wide range of OVA-peptide concentrations. We observed that while an optimal dose of peptide leads to cell cycle progression and proliferation, higher doses of peptide reduced cell growth, a phenomenon that was previously termed high-dose suppression. Our analysis of this phenomenon indicated that high-dose suppression is a consequence of cell cycle arrest, but not Fas-Fas ligand-dependent apoptosis or T-cell anergy, and that this growth arrest occurs in S phase, accompanied by reduced expression of CDK2 and cyclin A. Importantly, inhibition of MEK/ERK activation eliminated this growth suppression and cell cycle arrest, while it reduced the proliferative response to optimal antigenic stimulation. These results suggest that cell cycle arrest is the major mechanism regulating antigen-specific effector T-cell expansion, and that the MEK/ERK signaling pathway has both positive and negative effects, depending on the strength of antigenic stimulation.
Collapse
Affiliation(s)
- Shizuka Ohtsuka
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba 278-0022, Japan
| | - Shuhei Ogawa
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba 278-0022, Japan
| | - Ei Wakamatsu
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba 278-0022, Japan
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba 278-0022, Japan
| |
Collapse
|
18
|
Abstract
Natural killer cells are cytotoxic lymphocytes important in immune responses to cancer and multiple pathogens. However, chronic activation of NK cells can induce a hyporesponsive state. The molecular basis of the mechanisms underlying the generation and maintenance of this hyporesponsive condition are unknown, thus an easy and reproducible mechanism able to induce hyporesponsiveness on human NK cells would be very useful to gain understanding of this process. Human NK cells treated with ionomycin lose their ability to degranulate and secrete IFN-γ in response to a variety of stimuli, but IL-2 stimulation can compensate these defects. Apart from reductions in the expression of CD11a/CD18, no great changes were observed in the activating and inhibitory receptors expressed by these NK cells, however their transcriptional signature is different to that described for other hyporesponsive lymphocytes.
Collapse
|
19
|
Patakas A, Ji RR, Weir W, Connolly SE, Benson RA, Nadler SG, Brewer JM, McInnes IB, Garside P. Abatacept Inhibition of T Cell Priming in Mice by Induction of a Unique Transcriptional Profile That Reduces Their Ability to Activate Antigen-Presenting Cells. Arthritis Rheumatol 2016; 68:627-38. [DOI: 10.1002/art.39470] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/01/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Agapitos Patakas
- University of Glasgow, College of Medical, Veterinary, and Life Sciences; Glasgow UK
| | - Rui-Ru Ji
- Bristol-Myers Squibb, Research and Development; Princeton New Jersey
| | - William Weir
- University of Glasgow, College of Medical, Veterinary, and Life Sciences; Glasgow UK
| | - Sean E. Connolly
- Bristol-Myers Squibb, Research and Development; Princeton New Jersey
| | - Robert A. Benson
- University of Glasgow, College of Medical, Veterinary, and Life Sciences; Glasgow UK
| | - Steven G. Nadler
- Bristol-Myers Squibb, Research and Development; Princeton New Jersey
| | - James M. Brewer
- University of Glasgow, College of Medical, Veterinary, and Life Sciences; Glasgow UK
| | - Iain B. McInnes
- University of Glasgow, College of Medical, Veterinary, and Life Sciences; Glasgow UK
| | - Paul Garside
- University of Glasgow, College of Medical, Veterinary, and Life Sciences; Glasgow UK
| |
Collapse
|
20
|
Ndrg1 is a T-cell clonal anergy factor negatively regulated by CD28 costimulation and interleukin-2. Nat Commun 2015; 6:8698. [PMID: 26507712 PMCID: PMC4846325 DOI: 10.1038/ncomms9698] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 09/21/2015] [Indexed: 11/09/2022] Open
Abstract
Induction of T-cell clonal anergy involves serial activation of transcription factors, including NFAT and Egr2/3. However, downstream effector mechanisms of these transcription factors are not fully understood yet. Here we identify Ndrg1 as an anergy factor induced by Egr2. Ndrg1 is upregulated by anergic signalling and maintained at high levels in resting anergic T cells. Overexpression of Ndrg1 mimics the anergic state and knockout of the gene prevents anergy induction. Interestingly, Ndrg1 is phosphorylated and degraded by CD28 signalling in a proteasome-dependent manner, explaining the costimulation dependence of anergy prevention. Similarly, IL-2 treatment of anergic T cells, under conditions that lead to the reversal of anergy, also induces Ndrg1 phosphorylation and degradation. Finally, older Ndrg1-deficient mice show T-cell hyperresponsiveness and Ndrg1-deficient T cells aggravate inducible autoimmune inflammation. Thus, Ndrg1 contributes to the maintenance of clonal anergy and inhibition of T-cell-mediated inflammation.
Collapse
|
21
|
Rodvold JJ, Mahadevan NR, Zanetti M. Immune modulation by ER stress and inflammation in the tumor microenvironment. Cancer Lett 2015; 380:227-36. [PMID: 26525580 DOI: 10.1016/j.canlet.2015.09.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/16/2015] [Accepted: 09/19/2015] [Indexed: 12/18/2022]
Abstract
It is now increasingly evident that the immune system represents a barrier to tumor emergence, growth, and recurrence. Although this idea was originally proposed almost 50 years ago as the "immune surveillance hypothesis", it is commonly recognized that, with few rare exceptions, tumor cells always prevail. Thus, one of the central unsolved paradoxes of tumor immunology is how a tumor escapes immune control, which is reflected in the lack of effective autochthonous or vaccine-induced anti-tumor T cell responses. In this review, we discuss the role of the endoplasmic reticulum (ER) stress response/unfolded protein response (UPR) in the immunomodulation of myeloid cells and T cells. Specifically, we will discuss how the tumor cell UPR polarizes myeloid cells in a cell-extrinsic manner, and how in turn, thus polarized myeloid cells negatively affect T cell activation and clonal expansion.
Collapse
Affiliation(s)
- Jeffrey J Rodvold
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0815
| | - Navin R Mahadevan
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0815
| | - Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0815.
| |
Collapse
|
22
|
Peske JD, Woods AB, Engelhard VH. Control of CD8 T-Cell Infiltration into Tumors by Vasculature and Microenvironment. Adv Cancer Res 2015. [PMID: 26216636 DOI: 10.1016/bs.acr.2015.05.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CD8 T-cells are a critical brake on the initial development of tumors. In established tumors, the presence of CD8 T-cells is correlated with a positive patient prognosis, although immunosuppressive mechanisms limit their effectiveness and they are rarely curative without manipulation. Cancer immunotherapies aim to shift the balance back to dominant antitumor immunity through antibody blockade of immunosuppressive signaling pathways, vaccination, and adoptive transfer of activated or engineered T-cells. These approaches have yielded striking responses in small subsets of patients with solid tumors, most notably those with melanoma. Importantly, the subset of patients who respond to vaccination or immunosuppression blockade therapies are those with CD8 T-cells present in the tumor prior to initiating therapy. While current adoptive cell therapy approaches can be dramatically effective, they require infusion of extremely large numbers of T-cells, but the number that actually infiltrates the tumor is very small. Thus, poor representation of CD8 T-cells in tumors is a fundamental hurdle to successful immunotherapy, over and above the well-established barrier of immunosuppression. In this review, we discuss the factors that determine whether immune cells are present in tumors, with a focus on the representation of cytotoxic CD8 T-cells. We emphasize the critically important role of tumor-associated vasculature as a gateway that enables the active infiltration of both effector and naïve CD8 T-cells that exert antitumor activity. We also discuss strategies to enhance the gateway function and extend the effectiveness of immunotherapies to a broader set of cancer patients.
Collapse
Affiliation(s)
- J David Peske
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Amber B Woods
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Victor H Engelhard
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.
| |
Collapse
|
23
|
Janik DK, Lee WT. Staphylococcal Enterotoxin B (SEB) Induces Memory CD4 T Cell Anergy in vivo and Impairs Recall Immunity to Unrelated Antigens. ACTA ACUST UNITED AC 2015; 6:1-8. [PMID: 26807307 PMCID: PMC4720977 DOI: 10.4172/2155-9899.1000346] [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] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Naïve and memory T cells can utilize unique regulatory pathways to promote protection but prevent self-reactivity. A bacterial superantigen SEB exploits unique TCR proximal signaling processes in memory CD4 T cells to induce clonal anergy. The aim of this study was to determine if SEB could antagonize memory CD4 T cells in vivo and whether there would be consequences on recall immune responses. We evaluated Ab responses to a T-dependent antigen as a measurement of memory T cell helper function. METHOD BALB/c mice were primed with TNP-RGG to elicit memory B cells and also immunized with an ovalbumin peptide to elicit memory helper T cells. Another group of TNP-RGG immunized mice were used as adoptive transfer recipients of exogenous DO11.10 memory T cells. Mice were challenged with TNP-OVA with or without prior administration of SEB. B cells secreting IgM or IgG TNP-specific Ab were enumerated by ELISPOT as indicators of primary versus secondary humoral immunity. RESULTS Comparing the SEB and non-SEB-treated groups, the SEB-treated group failed to produce TNP-specific IgG in response to challenge with TNP-OVA, even if they were previously immunized with OVA. All groups produced IgM, indicating that the primary Ab responses and naïve helper T cells were not impacted by SEB. SEB had no negative impact when DO11.10 × Fyn-/- memory T cells were used as donor cells. CONCLUSION The present study indicated that SEB selectively targeted memory CD4 T cells in vivo and prevented helper function. Consequently, recall humoral immunity was lost. The data are most consistent with in vivo T cell anergy as opposed to indirect suppression as elimination of Fyn kinase restored helper function. These data suggest that bacterial superantigens can impair post-vaccination memory cell responses to unrelated antigens via their ability to target Vb families and antagonize memory cell activation.
Collapse
Affiliation(s)
- David K Janik
- The Department of Biomedical Sciences, School of Public Health, The University at Albany, USA
| | - William T Lee
- The Department of Biomedical Sciences, School of Public Health, The University at Albany, USA; The Laboratory of Immunology, The Wadsworth Center, New York State Department of Health, USA
| |
Collapse
|
24
|
Perera J, Liu X, Zhou Y, Joseph NE, Meng L, Turner JR, Huang H. Insufficient autoantigen presentation and failure of tolerance in a mouse model of rheumatoid arthritis. ACTA ACUST UNITED AC 2014; 65:2847-56. [PMID: 23840022 DOI: 10.1002/art.38085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 07/02/2013] [Indexed: 01/09/2023]
Abstract
OBJECTIVE In the K/BxN mouse model of rheumatoid arthritis, T cells reactive for the self antigen glucose-6-phosphate isomerase (GPI) escape negative selection even though GPI expression is ubiquitous. We sought to determine whether insufficient GPI presentation could account for the failure of negative selection and for the development of arthritis. METHODS To increase the antigen presentation of GPI, we generated transgenic mice expressing a membrane-bound form of GPI (mGPI) and crossed them with K/BxN mice. A monoclonal antibody specific for the α-chain of the KRN T cell receptor was generated to examine the fate of GPI-specific T cells. RESULTS The mGPI-transgenic mice presented GPI more efficiently and showed a dramatic increase in negative selection and an inhibition of arthritis. Interestingly, thymic negative selection remained incomplete in these mice, and the escaped autoreactive T cells were anergic in the peripheral lymphoid organs, suggesting that enhanced antigen presentation also induces peripheral tolerance. Despite this apparent tolerance induction toward GPI, these mice developed a chronic wasting disease, characterized by colonic inflammation with epithelial dysplasia, as well as a dramatic reduction in Treg cells. CONCLUSION These data indicate that insufficient autoantigen expression or presentation results in defects of both central and peripheral tolerance in the K/BxN mice. Our findings also support the idea that insufficient autoantigen levels may underlie the development of autoimmunity.
Collapse
|
25
|
Abstract
One of the mechanisms that are in place to control the activation of mature T cells that bear self-reactive antigen receptors is anergy, a long-term state of hyporesponsiveness that is established in T cells in response to suboptimal stimulation. T cells receive signals that result not only from antigen recognition and costimulation but also from other sources, including cytokine receptors, inhibitory receptors or metabolic sensors. Integration of those signals will determine T cell fate. Under conditions that induce anergy, T cells activate a program of gene expression that leads to the production of proteins that block T cell receptor signaling and inhibit cytokine gene expression. In this review we will examine those signals that determine functional outcome following antigen encounter, review current knowledge of the factors that ensure signaling inhibition and epigenetic gene silencing in anergic cells and explore the mechanisms that lead to the reversal of anergy and the reacquisition of effector functions.
Collapse
Affiliation(s)
- Rut Valdor
- Department of Pathology. Albert Einstein College of Medicine. Bronx, NY. USA
| | - Fernando Macian
- Department of Pathology. Albert Einstein College of Medicine. Bronx, NY. USA
| |
Collapse
|
26
|
Zanetti M. Cell-extrinsic effects of the tumor unfolded protein response on myeloid cells and T cells. Ann N Y Acad Sci 2013; 1284:6-11. [PMID: 23651187 DOI: 10.1111/nyas.12103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tumor-infiltrating myeloid cells, macrophages, and dendritic cells (DCs) are key regulators of tumor immunity and growth. The origin of tumor-derived signals that instruct myeloid cells in the tumor microenvironment is only partially understood. The endoplasmic reticulum (ER) stress response, or unfolded protein response (UPR), provides survival advantages to tumor growth. However, the cell-extrinsic effects of the tumor UPR on immune cells have not been explored. Our laboratory recently showed that the tumor UPR can be transmitted by yet unidentified factor(s) to myeloid cells, macrophages, and DCs. ER stress transmission to receiver myeloid cells upregulates the production of proinflammatory cytokines, and contextually of arginase I, leading to a proinflammatory/suppressive phenotype. DCs imprinted by tumor-borne ER stress transmissible factor(s) have decreased cross-presentation of antigen and defective cross-priming, causing T cell activation without proliferation. When DCs imprinted by transmissible ER stress are admixed with tumor cells and injected in vivo, facilitation of tumor growth is observed. Thus, tumor-borne ER stress plays a hitherto unappreciated role at the tumor/immune interface that ultimately facilitates tumor growth.
Collapse
Affiliation(s)
- Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
27
|
Destabilization of peptide:MHC interaction induces IL-2 resistant anergy in diabetogenic T cells. J Autoimmun 2013; 44:82-90. [PMID: 23895744 DOI: 10.1016/j.jaut.2013.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 06/17/2013] [Accepted: 07/02/2013] [Indexed: 01/09/2023]
Abstract
Autoreactive T cells are responsible for inducing several autoimmune diseases, including type 1 diabetes. We have developed a strategy to induce unresponsiveness in these cells by destabilizing the peptide:MHC ligand recognized by the T cell receptor. By introducing amino acid substitutions into the immunogenic peptide at residues that bind to the MHC, the half life of the peptide:MHC complex is severely reduced, thereby resulting in abortive T cell activation and anergy. By treating a monoclonal diabetogenic T cell population with an MHC variant peptide, the cells are rendered unresponsive to the wild type ligand, as measured by both proliferation and IL-2 production. Stimulation of T cells with MHC variant peptides results in minimal Erk1/2 phosphorylation or cell division. Variant peptide stimulation effectively initiates a signaling program dominated by sustained tyrosine phosphatase activity, including elevated SHP-1 activity. These negative signaling events result in an anergic phenotype in which the T cells are not competent to signal through the IL-2 receptor, as evidenced by a lack of phospho-Stat5 upregulation and proliferation, despite high expression of the IL-2 receptor. This unique negative signaling profile provides a novel means to shut down the anti-self response.
Collapse
|
28
|
Cobbold SP, Waldmann H. Regulatory cells and transplantation tolerance. Cold Spring Harb Perspect Med 2013; 3:3/6/a015545. [PMID: 23732858 DOI: 10.1101/cshperspect.a015545] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transplantation tolerance is a continuing therapeutic goal, and it is now clear that a subpopulation of T cells with regulatory activity (Treg) that express the transcription factor foxp3 are crucial to this aspiration. Although reprogramming of the immune system to donor-specific transplantation tolerance can be readily achieved in adult mouse models, it has yet to be successfully translated in human clinical practice. This requires that we understand the fundamental mechanisms by which donor antigen-specific Treg are induced and function to maintain tolerance, so that we can target therapies to enhance rather than impede these regulatory processes. Our current understanding is that Treg act via numerous molecular mechanisms, and critical underlying components such as mTOR inhibition, are only now emerging.
Collapse
Affiliation(s)
- Stephen P Cobbold
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, United Kingdom.
| | | |
Collapse
|
29
|
Ferragine CE, Walls CD, Davies SJ. Modulation of innate antigen-presenting cell function by pre-patent schistosome infection. PLoS Negl Trop Dis 2013; 7:e2136. [PMID: 23556020 PMCID: PMC3605154 DOI: 10.1371/journal.pntd.0002136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 02/11/2013] [Indexed: 01/10/2023] Open
Abstract
Schistosomes are intravascular helminths that infect over 200 million people worldwide. Deposition of eggs by adult schistosomes stimulates Th2 responses to egg antigens and induces granulomatous pathology that is a hallmark of schistosome infection. Paradoxically, schistosomes require host immune function for their development and reproduction and for egress of parasite eggs from the host. To identify potential mechanisms by which immune cells might influence parasite development prior to the onset of egg production, we assessed immune function in mice infected with developing schistosomes. We found that pre-patent schistosome infection is associated with a loss of T cell responsiveness to other antigens and is due to a diminution in the ability of innate antigen-presenting cells to stimulate T cells. Diminution of stimulatory capacity by schistosome worms specifically affected CD11b+ cells and did not require concomitant adaptive responses. We could not find evidence for production of a diffusible inhibitor of T cells by innate cells from infected mice. Rather, inhibition of T cell responsiveness by accessory cells required cell contact and only occurred when cells from infected mice outnumbered competent APCs by more than 3∶1. Finally, we show that loss of T cell stimulatory capacity may in part be due to suppression of IL-12 expression during pre-patent schistosome infection. Modulation of CD4+ T cell and APC function may be an aspect of host immune exploitation by schistosomes, as both cell types influence parasite development during pre-patent schistosome infection. The disease schistosomiasis is caused by a parasitic blood fluke found mainly in the tropics and subtropics and affects over 200 million people worldwide. Using mice to model human schistosome infection, our previous studies showed that schistosome development in the infected host is linked to host immune function, such that parasite development is impaired in hosts with immunological deficiencies. CD4+ T cells and cells of the monocyte/macrophage lineage are two types of immune cells that are involved in modulating schistosome development. In this study, we examined immune function in mice infected with developing schistosomes, to gain insights into how immune cells might influence parasite development. We found evidence of broad-spectrum suppression of CD4+ T cell responses during early schistosome infection. We also show that the loss of T cell responsiveness is due to impairment of T cell stimulation by CD11b+ cells. These findings suggest that exploitation of CD4+ T cells and monocytes/macrophages by schistosomes may involve parasite modification of the function of these cells.
Collapse
Affiliation(s)
- Christine E. Ferragine
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Colleen D. Walls
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Stephen J. Davies
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
30
|
Abstract
T cell activation leads to dramatic shifts in cell metabolism to protect against pathogens and to orchestrate the action of other immune cells. Quiescent T cells require predominantly ATP-generating processes, whereas proliferating effector T cells require high metabolic flux through growth-promoting pathways. Further, functionally distinct T cell subsets require distinct energetic and biosynthetic pathways to support their specific functional needs. Pathways that control immune cell function and metabolism are intimately linked, and changes in cell metabolism at both the cell and system levels have been shown to enhance or suppress specific T cell functions. As a result of these findings, cell metabolism is now appreciated as a key regulator of T cell function specification and fate. This review discusses the role of cellular metabolism in T cell development, activation, differentiation, and function to highlight the clinical relevance and opportunities for therapeutic interventions that may be used to disrupt immune pathogenesis.
Collapse
Affiliation(s)
- Nancie J MacIver
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
31
|
Mahadevan NR, Anufreichik V, Rodvold JJ, Chiu KT, Sepulveda H, Zanetti M. Cell-extrinsic effects of tumor ER stress imprint myeloid dendritic cells and impair CD8⁺ T cell priming. PLoS One 2012; 7:e51845. [PMID: 23272178 PMCID: PMC3525659 DOI: 10.1371/journal.pone.0051845] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 11/06/2012] [Indexed: 11/18/2022] Open
Abstract
Tumor-infiltrating myeloid cells, such as dendritic cells (BMDC), are key regulators of tumor growth. However, the tumor-derived signals polarizing BMDC to a phenotype that subverts cell-mediated anti-tumor immunity have yet to be fully elucidated. Addressing this unresolved problem we show that the tumor unfolded protein response (UPR) can function in a cell-extrinsic manner via the transmission of ER stress (TERS) to BMDC. TERS-imprinted BMDC upregulate the production of pro-inflammatory, tumorigenic cytokines but also the immunosuppressive enzyme arginase. Importantly, they downregulate cross-presentation of high-affinity antigen and fail to effectively cross-prime CD8(+) T cells, causing T cell activation without proliferation and similarly dominantly suppress cross-priming by bystander BMDC. Lastly, TERS-imprinted BMDC facilitate tumor growth in vivo with fewer tumor-infiltrating CD8(+) T cells. In sum, we demonstrate that tumor-borne ER stress imprints ab initio BMDC to a phenotype that recapitulates several of the inflammatory/suppressive characteristics ascribed to tumor-infiltrating myeloid cells, highlighting the tumor UPR as a critical controller of anti-tumor immunity and a new target for immune modulation in cancer.
Collapse
Affiliation(s)
- Navin R. Mahadevan
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Veronika Anufreichik
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Jeffrey J. Rodvold
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Kevin T. Chiu
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | | | - Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Microparticles bearing encephalitogenic peptides induce T-cell tolerance and ameliorate experimental autoimmune encephalomyelitis. Nat Biotechnol 2012; 30:1217-24. [PMID: 23159881 DOI: 10.1038/nbt.2434] [Citation(s) in RCA: 310] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 10/30/2012] [Indexed: 12/19/2022]
Abstract
Aberrant T-cell activation underlies many autoimmune disorders, yet most attempts to induce T-cell tolerance have failed. Building on previous strategies for tolerance induction that exploited natural mechanisms for clearing apoptotic debris, we show that antigen-decorated microparticles (500-nm diameter) induce long-term T-cell tolerance in mice with relapsing experimental autoimmune encephalomyelitis. Specifically, intravenous infusion of either polystyrene or biodegradable poly(lactide-co-glycolide) microparticles bearing encephalitogenic peptides prevents the onset and modifies the course of the disease. These beneficial effects require microparticle uptake by marginal zone macrophages expressing the scavenger receptor MARCO and are mediated in part by the activity of regulatory T cells, abortive T-cell activation and T-cell anergy. Together these data highlight the potential for using microparticles to target natural apoptotic clearance pathways to inactivate pathogenic T cells and halt the disease process in autoimmunity.
Collapse
|
33
|
Dudek TE, No DC, Seung E, Vrbanac VD, Fadda L, Bhoumik P, Boutwell CL, Power KA, Gladden AD, Battis L, Mellors EF, Tivey TR, Gao X, Altfeld M, Luster AD, Tager AM, Allen TM. Rapid evolution of HIV-1 to functional CD8⁺ T cell responses in humanized BLT mice. Sci Transl Med 2012; 4:143ra98. [PMID: 22814851 PMCID: PMC3685142 DOI: 10.1126/scitranslmed.3003984] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of mouse/human chimeras through the engraftment of human immune cells and tissues into immunodeficient mice, including the recently described humanized BLT (bone marrow, liver, thymus) mouse model, holds great promise to facilitate the in vivo study of human immune responses. However, little data exist regarding the extent to which cellular immune responses in humanized mice accurately reflect those seen in humans. We infected humanized BLT mice with HIV-1 as a model pathogen and characterized HIV-1-specific immune responses and viral evolution during the acute phase of infection. HIV-1-specific CD8(+) T cell responses in these mice were found to closely resemble those in humans in terms of their specificity, kinetics, and immunodominance. Viral sequence evolution also revealed rapid and highly reproducible escape from these responses, mirroring the adaptations to host immune pressures observed during natural HIV-1 infection. Moreover, mice expressing the protective HLA-B*57 allele exhibited enhanced control of viral replication and restricted the same CD8(+) T cell responses to conserved regions of HIV-1 Gag that are critical to its control of HIV-1 in humans. These data reveal that the humanized BLT mouse model appears to accurately recapitulate human pathogen-specific cellular immunity and the fundamental immunological mechanisms required to control a model human pathogen, aspects critical to the use of a small-animal model for human pathogens.
Collapse
Affiliation(s)
| | - Daniel C. No
- Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA, USA
| | - Edward Seung
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Vladimir D. Vrbanac
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Lena Fadda
- Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA, USA
| | | | | | - Karen A. Power
- Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA, USA
| | | | - Laura Battis
- Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA, USA
| | | | - Trevor R. Tivey
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Xiaojiang Gao
- Cancer and Immunology Program, Laboratory of Experimental Immunology, SAIC Frederick, NCI Frederick, Frederick, MD, USA
| | - Marcus Altfeld
- Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA, USA
| | - Andrew D. Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Andrew M. Tager
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Todd M. Allen
- Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA, USA
| |
Collapse
|
34
|
Akirav EM, Preston-Hurlburt P, Garyu J, Henegariu O, Clynes R, Schmidt AM, Herold KC. RAGE expression in human T cells: a link between environmental factors and adaptive immune responses. PLoS One 2012; 7:e34698. [PMID: 22509345 PMCID: PMC3324532 DOI: 10.1371/journal.pone.0034698] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 03/08/2012] [Indexed: 12/18/2022] Open
Abstract
The Receptor for Advanced Glycation Endproducts (RAGE) is a scavenger ligand that binds glycated endproducts as well as molecules released during cell death such as S100b and HMGB1. RAGE is expressed on antigen presenting cells where it may participate in activation of innate immune responses but its role in adaptive human immune responses has not been described. We have found that RAGE is expressed intracellularly in human T cells following TCR activation but constitutively on T cells from patients with diabetes. The levels of RAGE on T cells from patients with diabetes are not related to the level of glucose control. It co-localizes to the endosomes. Its expression increases in activated T cells from healthy control subjects but bystander cells also express RAGE after stimulation of the antigen specific T cells. RAGE ligands enhance RAGE expression. In patients with T1D, the level of RAGE expression decreases with T cell activation. RAGE+ T cells express higher levels of IL-17A, CD107a, and IL-5 than RAGE− cells from the same individual with T1D. Our studies have identified the expression of RAGE on adaptive immune cells and a role for this receptor and its ligands in modulating human immune responses.
Collapse
Affiliation(s)
- Eitan M. Akirav
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Paula Preston-Hurlburt
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Justin Garyu
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Octavian Henegariu
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Raphael Clynes
- Department of Medicine, Columbia University, New York, New York, United States of America
| | - Ann Marie Schmidt
- Department of Medicine, New York University, New York, New York, United States of America
| | - Kevan C. Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
35
|
Raϊch-Regué D, Grau-López L, Naranjo-Gómez M, Ramo-Tello C, Pujol-Borrell R, Martínez-Cáceres E, Borràs FE. Stable antigen-specific T-cell hyporesponsiveness induced by tolerogenic dendritic cells from multiple sclerosis patients. Eur J Immunol 2012; 42:771-82. [DOI: 10.1002/eji.201141835] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
36
|
Puddu P, Latorre D, Carollo M, Catizone A, Ricci G, Valenti P, Gessani S. Bovine lactoferrin counteracts Toll-like receptor mediated activation signals in antigen presenting cells. PLoS One 2011; 6:e22504. [PMID: 21799877 PMCID: PMC3143167 DOI: 10.1371/journal.pone.0022504] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 06/27/2011] [Indexed: 01/17/2023] Open
Abstract
Lactoferrin (LF), a key element in mammalian immune system, plays pivotal roles in host defence against infection and excessive inflammation. Its protective effects range from direct antimicrobial activities against a large panel of microbes, including bacteria, viruses, fungi and parasites, to antinflammatory and anticancer activities. In this study, we show that monocyte-derived dendritic cells (MD-DCs) generated in the presence of bovine LF (bLF) fail to undergo activation by up-modulating CD83, co-stimulatory and major histocompatibility complex molecules, and cytokine/chemokine secretion. Moreover, these cells are weak activators of T cell proliferation and retain antigen uptake activity. Consistent with an impaired maturation, bLF-MD-DC primed T lymphocytes exhibit a functional unresponsiveness characterized by reduced expression of CD154 and impaired expression of IFN-γ and IL-2. The observed imunosuppressive effects correlate with an increased expression of molecules with negative regulatory functions (i.e. immunoglobulin-like transcript 3 and programmed death ligand 1), indoleamine 2,3-dioxygenase, and suppressor of cytokine signaling-3. Interestingly, bLF-MD-DCs produce IL-6 and exhibit constitutive signal transducer and activator of transcription 3 activation. Conversely, bLF exposure of already differentiated MD-DCs completely fails to induce IL-6, and partially inhibits Toll-like receptor (TLR) agonist-induced activation. Cell-specific differences in bLF internalization likely account for the distinct response elicited by bLF in monocytes versus immature DCs, providing a mechanistic base for its multiple effects. These results indicate that bLF exerts a potent anti-inflammatory activity by skewing monocyte differentiation into DCs with impaired capacity to undergo activation and to promote Th1 responses. Overall, these bLF-mediated effects may represent a strategy to block excessive DC activation upon TLR-induced inflammation, adding further evidence for a critical role of bLF in directing host immune function.
Collapse
Affiliation(s)
- Patrizia Puddu
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Daniela Latorre
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Carollo
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Angela Catizone
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza, University of Rome, Rome, Italy
| | - Giulia Ricci
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Piera Valenti
- Department of Public Health Sciences and Infectious Diseases, Sapienza, University of Rome, Rome, Italy
| | - Sandra Gessani
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
- * E-mail:
| |
Collapse
|
37
|
Saeki K, Iwasa Y. T cell anergy as a strategy to reduce the risk of autoimmunity. J Theor Biol 2011; 277:74-82. [PMID: 21354182 DOI: 10.1016/j.jtbi.2011.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 01/18/2011] [Accepted: 02/16/2011] [Indexed: 10/18/2022]
Abstract
Some self-reactive immature T cells escape negative selection in the thymus and may cause autoimmune diseases later. In the periphery, if T cells are stimulated insufficiently by peptide-major histocompatibility complex, they become inactive and their production of cytokines changes, a phenomenon called "T cell anergy". In this paper, we explore the hypothesis that T cell anergy may function to reduce the risk of autoimmunity. The underlying logic is as follows: Since those self-reactive T cells that receive strong stimuli from self-antigens are eliminated in the thymus, T cells that receive strong stimuli in the periphery are likely to be non-self-reactive. As a consequence, when a T cell receives a weak stimulus, the likelihood that the cell is self-reactive is higher than in the case that it receives a strong stimulus. Therefore, inactivation of the T cell may reduce the danger of autoimmunity. We consider the formalism in which each T cell chooses its response depending on the strength of stimuli in order to reduce the risk of autoimmune diseases while maintaining its ability to attack non-self-antigens effectively. The optimal T cell responses to a weak and a strong stimulus are obtained both when the cells respond in a deterministic manner and when they respond in a probabilistic manner. We conclude that T cell anergy is the optimal response when a T cell meets with antigen-presenting cells many times in its lifetime, and when the product of the autoimmunity risk and the number of self-reactive T cells has an intermediate value.
Collapse
Affiliation(s)
- Koichi Saeki
- Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka 812-8581, Japan.
| | | |
Collapse
|
38
|
Tohn R, Blumenfeld H, Haeryfar SMM, Veerapen N, Besra GS, Porcelli SA, Delovitch TL. Stimulation of a shorter duration in the state of anergy by an invariant natural killer T cell agonist enhances its efficiency of protection from type 1 diabetes. Clin Exp Immunol 2011; 164:26-41. [PMID: 21361909 DOI: 10.1111/j.1365-2249.2011.04323.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We have reported previously that treatment of non-obese diabetic (NOD) mice with the invariant natural killer T (iNK T) cell agonist α-galactosylceramide C26:0 (α-GalCer) or its T helper type 2 (Th2)-biasing derivative α-GalCer C20:2 (C20:2) protects against type 1 diabetes (T1D), with C20:2 yielding greater protection. After an initial response to α-GalCer, iNK T cells become anergic upon restimulation. While such anergic iNK T cells can induce tolerogenic dendritic cells (DCs) that mediate protection from T1D, chronic administration of α-GalCer also results in long-lasting anergy accompanied by significantly reduced iNK T cell frequencies, which raises concerns about its long-term therapeutic use. In this study, our objective was to understand more clearly the roles of anergy and induction of tolerogenic DCs in iNK T cell-mediated protection from T1D and to circumvent potential complications associated with α-GalCer. We demonstrate that NOD iNK T cells activated during multi-dose (MD) treatment in vivo with C20:2 enter into and exit from anergy more rapidly than after activation by α-GalCer. Importantly, this shorter duration of iNK T cells in the anergic state promotes the more rapid induction of tolerogenic DCs and reduced iNK T cell death, and enables C20:2 stimulated iNK T cells to elicit enhanced protection from T1D. Our findings further that suggest C20:2 is a more effective therapeutic drug than α-GalCer for protection from T1D. Moreover, the characteristics of C20:2 provide a basis of selection of next-generation iNK T cell agonists for the prevention of T1D.
Collapse
Affiliation(s)
- R Tohn
- Laboratory of Autoimmune Diabetes, Robarts Research Institute Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
39
|
Ponchel F, Cuthbert RJ, Goëb V. IL-7 and lymphopenia. Clin Chim Acta 2010; 412:7-16. [PMID: 20850425 DOI: 10.1016/j.cca.2010.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 09/01/2010] [Accepted: 09/01/2010] [Indexed: 01/10/2023]
Abstract
Interleukin-7 (IL-7) is a growth and anti-apoptotic factor for T-lymphocytes, with potential for clinical use in the treatment of immunodeficiencies due to loss of T-cells. Lymphopenia induced by disease (HIV infection, hemodialysis or Idiopathic CD4+ lymphopenia) or by treatment (high dose chemotherapy or depleting antibodies) for cancer or auto-immune diseases results in increased circulating levels of IL-7 which decline with T-cell recovery, however, the mechanism of such response remains to be elucidated. Furthermore, IL-7 is a major player in the regulation of peripheral T-cell homeostasis and as such is an important candidate cytokine for therapy aimed at improving T-cell reconstitution following lymphopenia. Anti- IL-7 is on the other hand proposed to treat conditions where IL-7 may play a more direct role in pathogenesis such as autoimmune disease like Rheumatoid Arthritis, Multiple Sclerosis or Inflammatory Bowel disease.
Collapse
Affiliation(s)
- Frederique Ponchel
- Leeds Institute of Molecular Medicine, Section of Musculoskeletal disease, the University of Leeds, Leeds, UK.
| | | | | |
Collapse
|
40
|
Ramanathan S, Dubois S, Gagnon J, Leblanc C, Mariathasan S, Ferbeyre G, Rottapel R, Ohashi PS, Ilangumaran S. Regulation of cytokine-driven functional differentiation of CD8 T cells by suppressor of cytokine signaling 1 controls autoimmunity and preserves their proliferative capacity toward foreign antigens. THE JOURNAL OF IMMUNOLOGY 2010; 185:357-66. [PMID: 20519645 DOI: 10.4049/jimmunol.1000066] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have previously shown that naive CD8 T cells exposed to IL-7 or IL-15 in the presence of IL-21 undergo Ag-independent proliferation with concomitant increase in TCR sensitivity. In this study, we examined whether CD8 T cells that accumulate in suppressor of cytokine signaling 1 (SOCS1)-deficient mice because of increased IL-15 signaling in vivo would respond to an autoantigen expressed at a very low level using a mouse model of autoimmune diabetes. In this model, P14 TCR transgenic CD8 T cells (P14 cells) adoptively transferred to rat insulin promoter-glycoprotein (RIP-GP) mice, which express the cognate Ag in the islets, do not induce diabetes unless the donor cells are stimulated by exogenous Ag. Surprisingly, SOCS1-deficient P14 cells, which expanded robustly following IL-15 stimulation, proliferated poorly in response to Ag and failed to cause diabetes in RIP-GP mice. SOCS1-deficient CD8 T cells expressing a polyclonal TCR repertoire also showed defective expansion following in vivo Ag stimulation. Notwithstanding the Ag-specific proliferation defect, SOCS1-null P14 cells produced IFN-gamma and displayed potent cytolytic activity upon Ag stimulation, suggesting that SOCS1-null CD8 T cells underwent cytokine-driven functional differentiation that selectively compromised their proliferative response to Ag but not to cytokines. Cytokine-driven homeostatic expansion in lymphopenic RIP-GP mice allowed SOCS1-null, but not wild-type, P14 cells to exert their pathogenic potential even without Ag stimulation. These findings suggest that by attenuating cytokine-driven proliferation and functional differentiation, SOCS1 not only controls the pathogenicity of autoreactive cells but also preserves the ability of CD8 T cells to proliferate in response to Ags.
Collapse
Affiliation(s)
- Sheela Ramanathan
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Regulatory T cells (Treg cells) play critical roles in the induction of peripheral tolerance to self- and foreign antigens. Naturally occurring CD4(+)CD25(+) Treg cells, which characteristically express the transcription factor forkhead box protein P3 (Foxp3), have been studied intensively because their deficiency abrogates self-tolerance and causes autoimmune disease. However, several lines of evidence suggest that additional important mechanisms other than the Foxp3 system are required to enforce immunological self-tolerance in the periphery. Interleukin-10 (IL-10) is a regulatory cytokine that plays a central role in controlling inflammatory processes, and IL-10-secreting T cells may constitute an additional mechanism that are responsible for peripheral tolerance. Type-1 T regulatory (Tr1) cells, CD46-stimulated IL-10-secreting T cells, and IL-10-secreting T cells induced by vitamin D3 (VitD3) and dexamethasone (Dex) are induced populations with significant regulatory activities. However, assessing the detailed physiological function of these cells is difficult, because of the lack of specific markers that can reliably differentiate the population of IL-10-secreting Treg cells from other T cells. Recently, CD4(+)CD25(-)LAP(+) T cells, CD4(+)NKG2D(+) T cells, CD4(+)IL-7R(-) T cells, and CD4(+)CD25(-)LAG3(+) T cells have been reported as naturally present IL-10-secreting Treg cells. Although the relationship between these induced and naturally present IL-10-secreting Treg cells is unclear, elucidation of their respective roles in modulating immune responses is crucial to understand T cell-mediated tolerance. Furthermore, the identification of specific markers and molecular signatures will enable the purification or induction of IL-10-secreting Treg cells for the treatment of patients having inflammatory diseases.
Collapse
Affiliation(s)
- Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
42
|
Kelly M, Bharadwaj AS, Tacke F, Chao H. Regulatory T cells and immune tolerance to coagulation factor IX in the context of intramuscular AAV1 gene transfer. Mol Ther 2009; 18:361-9. [PMID: 19935782 DOI: 10.1038/mt.2009.269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Regulatory T cells play a major role in induction and maintenance of immune tolerance and immunological homeostasis. A variety of strategies have been attempted to induce regulatory T cells for control of unwanted, adverse immunity in autoimmune diseases, transplantation as well as gene transfer. We recently reported efficient induction of immune tolerance to coagulation factor IX (FIX) following intramuscular AAV1 gene transfer. In the current study, we performed a systematic and comprehensive examination of the role and function of regulatory T cells in induction and maintenance of FIX tolerance in the context of intramuscular AAV1 gene transfer. We observed no significant upregulation of regulatory T cells in the FIX-tolerant mice. In addition, adoptive transfer of splenocytes from FIX-tolerant mice did not suppress anti-hFIX immunity in recipient mice. Both in vitro and in vivo depletion of regulatory T cells failed to reverse FIX tolerance. These observations revealed that regulatory T cells do not play a significant role in the maintenance/protection of the established FIX tolerance. Our results provide critical insight into the role and function of regulatory T cells in induction and maintenance/protection of immune tolerance in gene transfer, complementing the current paradigm of immune tolerance mechanism.
Collapse
Affiliation(s)
- Meagan Kelly
- Division of Hematology/Oncology, Cancer Institute, Department of Medicine, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | |
Collapse
|
43
|
Lai YP, Lin CC, Liao WJ, Tang CY, Chen SC. CD4+ T cell-derived IL-2 signals during early priming advances primary CD8+ T cell responses. PLoS One 2009; 4:e7766. [PMID: 19901991 PMCID: PMC2770320 DOI: 10.1371/journal.pone.0007766] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 09/30/2009] [Indexed: 01/22/2023] Open
Abstract
Stimulating naïve CD8+ T cells with specific antigens and costimulatory signals is insufficient to induce optimal clonal expansion and effector functions. In this study, we show that the activation and differentiation of CD8+ T cells require IL-2 provided by activated CD4+ T cells at the initial priming stage within 0–2.5 hours after stimulation. This critical IL-2 signal from CD4+ cells is mediated through the IL-2Rβγ of CD8+ cells, which is independent of IL-2Rα. The activation of IL-2 signaling advances the restriction point of the cell cycle, and thereby expedites the entry of antigen-stimulated CD8+ T-cell into the S phase. Besides promoting cell proliferation, IL-2 stimulation increases the amount of IFNγ and granzyme B produced by CD8+ T cells. Furthermore, IL-2 at priming enhances the ability of P14 effector cells generated by antigen activation to eradicate B16.gp33 tumors in vivo. Therefore, our studies demonstrate that a full CD8+ T-cell response is elicited by a critical temporal function of IL-2 released from CD4+ T cells, providing mechanistic insights into the regulation of CD8+ T cell activation and differentiation.
Collapse
Affiliation(s)
- Yo-Ping Lai
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Ching Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Jung Liao
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Yung Tang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Ching Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
44
|
Zheng Y, Delgoffe GM, Meyer CF, Chan W, Powell JD. Anergic T cells are metabolically anergic. THE JOURNAL OF IMMUNOLOGY 2009; 183:6095-101. [PMID: 19841171 DOI: 10.4049/jimmunol.0803510] [Citation(s) in RCA: 215] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Full T cell activation requires TCR engagement (signal 1) in the context of costimulation (signal 2). Costimulation is required for maximal expression of effector cytokines and prevention of T cell anergy. It has become increasingly clear that another major function of costimulation is to up-regulate the metabolic machinery necessary for T cell function. In this report we demonstrate that anergic T cells are metabolically anergic, in that upon full stimulation (signals 1 plus 2) they fail to up-regulate the machinery necessary to support increased metabolism. These findings suggest that one mechanism responsible for the maintenance of T cell anergy is failure to up-regulate the metabolic machinery. Furthermore, we demonstrate that by blocking leucine, glucose, and energy metabolism, T cell activation is mitigated. Additionally, inhibition of these metabolic pathways during T cell activation leads to anergy in Th1-differentiated cells. Overall, our findings extend the role of T cell metabolism in regulating T cell function.
Collapse
Affiliation(s)
- Yan Zheng
- The Sidney-Kimmel Cancer Research Center, The Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
45
|
Malhotra I, Dent A, Mungai P, Wamachi A, Ouma JH, Narum DL, Muchiri E, Tisch DJ, King CL. Can prenatal malaria exposure produce an immune tolerant phenotype? A prospective birth cohort study in Kenya. PLoS Med 2009; 6:e1000116. [PMID: 19636353 PMCID: PMC2707618 DOI: 10.1371/journal.pmed.1000116] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 06/17/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Malaria in pregnancy can expose the fetus to malaria-infected erythrocytes or their soluble products, thereby stimulating T and B cell immune responses to malaria blood stage antigens. We hypothesized that fetal immune priming, or malaria exposure in the absence of priming (putative tolerance), affects the child's susceptibility to subsequent malaria infections. METHODS AND FINDINGS We conducted a prospective birth cohort study of 586 newborns residing in a malaria-holoendemic area of Kenya who were examined biannually to age 3 years for malaria infection, and whose malaria-specific cellular and humoral immune responses were assessed. Newborns were classified as (i) sensitized (and thus exposed), as demonstrated by IFNgamma, IL-2, IL-13, and/or IL-5 production by cord blood mononuclear cells (CBMCs) to malaria blood stage antigens, indicative of in utero priming (n = 246), (ii) exposed not sensitized (mother Plasmodium falciparum [Pf]+ and no CBMC production of IFNgamma, IL-2, IL-13, and/or IL-5, n = 120), or (iii) not exposed (mother Pf-, no CBMC reactivity, n = 220). Exposed not sensitized children had evidence for prenatal immune experience demonstrated by increased IL-10 production and partial reversal of malaria antigen-specific hyporesponsiveness with IL-2+IL-15, indicative of immune tolerance. Relative risk data showed that the putatively tolerant children had a 1.61 (95% confidence interval [CI] 1.10-2.43; p = 0.024) and 1.34 (95% CI 0.95-1.87; p = 0.097) greater risk for malaria infection based on light microscopy (LM) or PCR diagnosis, respectively, compared to the not-exposed group, and a 1.41 (95%CI 0.97-2.07, p = 0.074) and 1.39 (95%CI 0.99-2.07, p = 0.053) greater risk of infection based on LM or PCR diagnosis, respectively, compared to the sensitized group. Putatively tolerant children had an average of 0.5 g/dl lower hemoglobin levels (p = 0.01) compared to the other two groups. Exposed not sensitized children also had 2- to 3-fold lower frequency of malaria antigen-driven IFNgamma and/or IL-2 production (p<0.001) and higher IL-10 release (p<0.001) at 6-month follow-ups, when compared to sensitized and not-exposed children. Malaria blood stage-specific IgG antibody levels were similar among the three groups. CONCLUSIONS These results show that a subset of children exposed to malaria in utero acquire a tolerant phenotype to blood-stage antigens that persists into childhood and is associated with an increased susceptibility to malaria infection and anemia. This finding could have important implications for malaria vaccination of children residing in endemic areas.
Collapse
MESH Headings
- Adult
- Animals
- Antibodies, Protozoan/blood
- Antigens, Protozoan/blood
- Antigens, Protozoan/immunology
- Antigens, Protozoan/metabolism
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Female
- Fetal Blood/immunology
- Humans
- Immune Tolerance
- Infant, Newborn
- Kenya/epidemiology
- Malaria, Falciparum/epidemiology
- Malaria, Falciparum/immunology
- Male
- Maternal-Fetal Exchange/immunology
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Merozoite Surface Protein 1/immunology
- Merozoite Surface Protein 1/metabolism
- Plasmodium falciparum
- Pregnancy
- Pregnancy Complications, Parasitic/epidemiology
- Pregnancy Complications, Parasitic/immunology
- Prospective Studies
- Protozoan Proteins/immunology
- Protozoan Proteins/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/parasitology
Collapse
Affiliation(s)
- Indu Malhotra
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Arlene Dent
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Peter Mungai
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
- Division of Vector Borne Diseases, Nairobi, Kenya
| | - Alex Wamachi
- Kenya Medical Research Institute, Nairobi, Kenya
| | - John H. Ouma
- Division of Vector Borne Diseases, Nairobi, Kenya
| | - David L. Narum
- Malaria Vaccine Development Unit, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Eric Muchiri
- Division of Vector Borne Diseases, Nairobi, Kenya
| | - Daniel J. Tisch
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Christopher L. King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
- Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
46
|
Abstract
The potent immunosuppressive action of rapamycin is commonly ascribed to inhibition of growth factor-induced T cell proliferation. However, it is now evident that the serine/threonine protein kinase mammalian target of rapamycin (mTOR) has an important role in the modulation of both innate and adaptive immune responses. mTOR regulates diverse functions of professional antigen-presenting cells, such as dendritic cells (DCs), and has important roles in the activation of effector T cells and the function and proliferation of regulatory T cells. In this Review, we discuss our current understanding of the mTOR pathway and the consequences of mTOR inhibition, both in DCs and T cells, including new data on the regulation of forkhead box P3 expression.
Collapse
|
47
|
Satpute SR, Rajaiah R, Polumuri SK, Moudgil KD. Tolerization with Hsp65 induces protection against adjuvant-induced arthritis by modulating the antigen-directed interferon-gamma, interleukin-17, and antibody responses. ACTA ACUST UNITED AC 2009; 60:103-13. [PMID: 19116924 DOI: 10.1002/art.24139] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Pretreatment of Lewis rats with soluble mycobacterial Hsp65 affords protection against subsequent adjuvant-induced arthritis (AIA). This study was aimed at unraveling the mechanisms underlying mycobacterial Hsp65-induced protection against arthritis, using contemporary parameters of immunity. METHODS Lewis rats were given 3 intraperitoneal injections of mycobacterial Hsp65 in solution prior to the initiation of AIA with heat-killed Mycobacterium tuberculosis. Thereafter, mycobacterial Hsp65-specific T cell proliferative, cytokine, and antibody responses were tested in tolerized rats. The roles of anergy and the indoleamine 2,3 dioxygenase (IDO)-tryptophan pathway in tolerance induction were assessed, and the frequency and suppressive function of CD4+FoxP3+ Treg cells were monitored. Also tested was the effect of mycobacterial Hsp65 tolerization on T cell responses to AIA-related mycobacterial Hsp70, mycobacterial Hsp10, and rat Hsp65. RESULTS The AIA-protective effect of mycobacterial Hsp65-induced tolerance was associated with a significantly reduced T cell proliferative response to mycobacterial Hsp65, which was reversed by interleukin-2 (IL-2), indicating anergy induction. The production of interferon-gamma (but not IL-4/IL-10) was increased, with concurrent down-regulation of IL-17 expression by mycobacterial Hsp65-primed T cells. Neither the frequency nor the suppressive activity of CD4+FoxP3+ T cells changed following tolerization, but the serum level of anti-mycobacterial Hsp65 antibodies was increased. However, no evidence was observed for a role of IDO or cross-tolerance to mycobacterial Hsp70, mycobacterial Hsp10, or rat Hsp65. CONCLUSION Tolerization with soluble mycobacterial Hsp65 leads to suppression of IL-17, anergy induction, and enhanced production of anti-mycobacterial Hsp65 antibodies, which play a role in protection against AIA. These results are relevant to the development of effective immunotherapeutic approaches for autoimmune arthritis.
Collapse
|
48
|
King JW, Thomas S, Corsi F, Gao L, Dina R, Gillmore R, Pigott K, Kaisary A, Stauss HJ, Waxman J. IL15 Can Reverse the Unresponsiveness of Wilms' Tumor Antigen-Specific CTL in Patients with Prostate Cancer. Clin Cancer Res 2009; 15:1145-54. [DOI: 10.1158/1078-0432.ccr-08-1821] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Khatri M, Sharma JM. Response of embryonic chicken lymphoid cells to infectious bursal disease virus. Vet Immunol Immunopathol 2009; 127:316-24. [DOI: 10.1016/j.vetimm.2008.10.327] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 10/16/2008] [Accepted: 10/20/2008] [Indexed: 11/16/2022]
|
50
|
Kawamura K, Yao K, Shukaliak-Quandt JA, Huh J, Baig M, Quigley L, Ito N, Necker A, McFarland HF, Muraro PA, Martin R, Ito K. Different development of myelin basic protein agonist- and antagonist-specific human TCR transgenic T cells in the thymus and periphery. THE JOURNAL OF IMMUNOLOGY 2008; 181:5462-72. [PMID: 18832703 DOI: 10.4049/jimmunol.181.8.5462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Myelin basic protein (MBP)-specific T cells are thought to play a role in the development of multiple sclerosis. MBP residues 111-129 compose an immunodominant epitope cluster restricted by HLA-DRB1*0401. The sequence of residues 111-129 of MBP (MBP(111-129)) differs in humans (MBP122:Arg) and mice (MBP122:Lys) at aa 122. We previously found that approximately 50% of human MBP(111-129) (MBP122:Arg)-specific T cell clones, including MS2-3C8 can proliferate in response to mouse MBP(111-129) (MBP122:Lys). However, the other half of T cell clones, including HD4-1C2, cannot proliferate in response to MBP(111-129) (MBP122:Lys). We found that MBP(111-129) (MBP122:Lys) is an antagonist for HD4-1C2 TCR, therefore, MS2-3C8 and HD4-1C2 TCRs are agonist- and antagonist-specific TCRs in mice, respectively. Therefore, we examined the development of HD4-1C2 TCR and MS2-3C8 TCR transgenic (Tg) T cells in the thymus and periphery. We found that dual TCR expression exclusively facilitates the development of MBP(111-129) TCR Tg T cells in the periphery of HD4-1C2 TCR/HLA-DRB1*0401 Tg mice although it is not required for their development in the thymus. We also found that MS2-3C8 TCR Tg CD8(+) T cells develop along with MS2-3C8 TCR Tg CD4(+) T cells, and that dual TCR expression was crucial for the development of MS2-3C8 TCR Tg CD4(+) and CD8(+) T cells in the thymus and periphery, respectively. These results suggest that thymic and peripheral development of MBP-specific T cells are different; however, dual TCR expression can facilitate their development.
Collapse
Affiliation(s)
- Kazuyuki Kawamura
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|