1
|
Sollid LM. Tolerance-inducing therapies in coeliac disease - mechanisms, progress and future directions. Nat Rev Gastroenterol Hepatol 2024; 21:335-347. [PMID: 38336920 DOI: 10.1038/s41575-024-00895-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
Coeliac disease is an autoinflammatory condition caused by immune reactions to cereal gluten proteins. Currently, the only available treatment for the condition is a lifelong avoidance of gluten proteins in the diet. There is an unmet need for alternative therapies. Coeliac disease has a strong association with certain HLA-DQ allotypes (DQ2.5, DQ2.2 and DQ8), and these disease-associated HLA-DQ molecules present deamidated gluten peptides to gluten-specific CD4+ T cells. The gluten-specific CD4+ T cells are the drivers of the immune reactions leading to coeliac disease. Once established, the clonotypes of gluten-specific CD4+ T cells persist for decades, explaining why patients must adhere to a gluten-free diet for life. Given the key pathogenic role of gluten-specific CD4+ T cells, tolerance-inducing therapies that target these T cells are attractive for treatment of the disorder. Lessons learned from coeliac disease might provide clues for treatment of other HLA-associated diseases for which the disease-driving antigens are unknown. Thus, intensive efforts have been and are currently implemented to bring an effective tolerance-inducing therapy for coeliac disease. This Review discusses mechanisms of the various approaches taken, summarizing the progress made, and highlights future directions in this field.
Collapse
Affiliation(s)
- Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
2
|
Külp M, Siemund AL, Larghero P, Dietz A, Alten J, Cario G, Eckert C, Caye-Eude A, Cavé H, Bardini M, Cazzaniga G, De Lorenzo P, Valsecchi MG, Diehl L, Bonig H, Meyer C, Marschalek R. The immune checkpoint ICOSLG is a relapse-predicting biomarker and therapeutic target in infant t(4;11) acute lymphoblastic leukemia. iScience 2022; 25:104613. [PMID: 35800767 PMCID: PMC9253708 DOI: 10.1016/j.isci.2022.104613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/14/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
The most frequent genetic aberration leading to infant ALL (iALL) is the chromosomal translocation t(4;11), generating the fusion oncogenes KMT2A:AFF1 and AFF1:KMT2A, respectively. KMT2A-r iALL displays a dismal prognosis through high relapse rates and relapse-associated mortality. Relapse occurs frequently despite ongoing chemotherapy and without the accumulation of secondary mutations. A rational explanation for the observed chemo-resistance and satisfactory treatment options remain to be elucidated. We found that elevated ICOSLG expression level at diagnosis was associated with inferior event free survival (EFS) in a cohort of 43 patients with t(4;-11) iALL and that a cohort of 18 patients with iALL at relapse displayed strongly increased ICOSLG expression. Furthermore, co-culturing t(4;11) ALL cells (ICOSLGhi) with primary T-cells resulted in the development of Tregs. This was impaired through treatment with a neutralizing ICOSLG antibody. These findings imply ICOSLG (1) as a relapse-predicting biomarker, and (2) as a therapeutic target involved in a potential immune evasion relapse-mechanism of infant t(4;11) ALL. Early growth response 3 (EGR3) is a direct transactivator of the immune checkpoint gene ICOSLG high ICOSLG expression at diagnosis is predictive for ALL relapse EGR3 and ICOSLG expressions are relapse-associated expression of ICOSLG on t(4;11) ALL cells leads to the rapid expansion of Tregs
Collapse
|
3
|
Richardson N, Wraith DC. Advancement of antigen-specific immunotherapy: knowledge transfer between allergy and autoimmunity. IMMUNOTHERAPY ADVANCES 2021; 1:ltab009. [PMID: 35919740 PMCID: PMC9327121 DOI: 10.1093/immadv/ltab009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/28/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Targeted restoration of immunological tolerance to self-antigens or innocuous environmental allergens represents the ultimate aim of treatment options in autoimmune and allergic disease. Antigen-specific immunotherapy (ASI) is the only intervention that has proven disease-modifying efficacy as evidenced by induction of long-term remission in a number of allergic conditions. Mounting evidence is now indicating that specific targeting of pathogenic T cells in autoinflammatory and autoimmune settings enables effective restoration of immune homeostasis between effector and regulatory cells and alters the immunological course of disease. Here, we discuss the key lessons learned during the development of antigen-specific immunotherapies and how these can be applied to inform future interventions. Armed with this knowledge and current high-throughput technology to track immune cell phenotype and function, it may no longer be a matter of ‘if’ but ‘when’ this ultimate aim of targeted tolerance restoration is realised.
Collapse
Affiliation(s)
- Naomi Richardson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - David Cameron Wraith
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
4
|
Wang W, Wu Q, Wang Z, Ren S, Shen H, Shi W, Xu Y. Development of a Prognostic Model for Ovarian Cancer Patients Based on Novel Immune Microenvironment Related Genes. Front Oncol 2021; 11:647273. [PMID: 33869044 PMCID: PMC8045757 DOI: 10.3389/fonc.2021.647273] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
Ovarian cancer (OV) has become the most lethal gynecological cancer. However, its treatment methods and staging system are far from ideal. In the present study, taking the advantage of large-scale public cohorts, we extracted a list of immune-related prognostic genes that differentially expressed in tumor and normal ovarian tissues. Importantly, an individualized immune-related gene based prognostic model (IPM) for OV patients were developed. Furthermore, we validated our IPM in Gene Expression Omnibus (GEO) repository and compared the immune landscape and pathways between high-risk and low-risk groups. The results of our study can serve as an important model to identify the immune subset of patients and has potential for use in immune therapeutic selection and patient management.
Collapse
Affiliation(s)
- Wei Wang
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China.,Department of Medicine, Nantong University Xinling College, Nantong, China
| | - Qianqian Wu
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China
| | - Ziheng Wang
- Department of Medicine, Nantong University Xinling College, Nantong, China
| | - Shiqi Ren
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China.,Department of Medicine, Nantong University Xinling College, Nantong, China
| | - Hanyu Shen
- Department of Medicine, Nantong University Xinling College, Nantong, China
| | - Wenyu Shi
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yunzhao Xu
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China.,Department of Obstetrics and Gynecology, Nantong University Affiliated Hospital, Nantong, China
| |
Collapse
|
5
|
Zhou X, Zhang FY, Liu Y, Wei DX. A Risk Prediction Model for Breast Cancer Based on Immune Genes Related to Early Growth Response Proteins Family. Front Mol Biosci 2021; 7:616547. [PMID: 33614706 PMCID: PMC7887293 DOI: 10.3389/fmolb.2020.616547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/21/2020] [Indexed: 11/19/2022] Open
Abstract
Early growth response proteins (EGRs), a transcriptional regulatory family comprised of EGR1, EGR2, EGR3, and EGR 4, are reportedly involved in a vast array of functions. However, EGRs, as a whole, are rarely studied in breast cancer cases. This research was performed based on public datasets. The results demonstrated that, except EGR4, the other EGRs were differentially expressed genes in breast cancer. Subsequently, this study determined the prognosis significance of the EGR family, higher expression levels of EGRs indicating better overall survival (OS) and disease-free survival (DFS), except EGR4. So we attempted to explore the potential mechanism behind the prognostic value of EGRs. At the DNA level, however, neither DNA methylation status nor genetic alterations of EGRs contributed to the prognosis significance. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that EGRs were involved in several immune-related functions. Afterward, we assessed the correlation between EGRs and the immune system before establishing a risk prediction model with a 14-gene immune signature associated with EGRs, a prognostic nomogram predicting individuals’ 1-, 3-, and 5-year survival probabilities. The risk score was an independent prognosis predictor in the breast cancer cohorts. This study evidenced EGRs’ significance for tumor immunity, demonstrating that the EGR family may be a potential immunotherapeutic target for breast cancer. The 14-gene immune signature is a promising prognostic biomarker in breast cancer.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Breast Surgery, Zibo Maternal and Child Health Hospital, Zibo, China
| | - Fang-Yuan Zhang
- Department of Breast Surgery, Zibo Maternal and Child Health Hospital, Zibo, China
| | - Yan Liu
- Department of Breast Surgery, Zibo Maternal and Child Health Hospital, Zibo, China
| | - Dong-Xin Wei
- Department of Breast Surgery, Zibo Maternal and Child Health Hospital, Zibo, China
| |
Collapse
|
6
|
Ambler R, Edmunds GL, Tan SL, Cirillo S, Pernes JI, Ruan X, Huete-Carrasco J, Wong CCW, Lu J, Ward J, Toti G, Hedges AJ, Dovedi SJ, Murphy RF, Morgan DJ, Wülfing C. PD-1 suppresses the maintenance of cell couples between cytotoxic T cells and target tumor cells within the tumor. Sci Signal 2020; 13:13/649/eaau4518. [PMID: 32934075 DOI: 10.1126/scisignal.aau4518] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The killing of tumor cells by CD8+ T cells is suppressed by the tumor microenvironment, and increased expression of inhibitory receptors, including programmed cell death protein-1 (PD-1), is associated with tumor-mediated suppression of T cells. To find cellular defects triggered by tumor exposure and associated PD-1 signaling, we established an ex vivo imaging approach to investigate the response of antigen-specific, activated effector CD8+ tumor-infiltrating lymphocytes (TILs) after interaction with target tumor cells. Although TIL-tumor cell couples readily formed, couple stability deteriorated within minutes. This was associated with impaired F-actin clearing from the center of the cellular interface, reduced Ca2+ signaling, increased TIL locomotion, and impaired tumor cell killing. The interaction of CD8+ T lymphocytes with tumor cell spheroids in vitro induced a similar phenotype, supporting a critical role of direct T cell-tumor cell contact. Diminished engagement of PD-1 within the tumor, but not acute ex vivo blockade, partially restored cell couple maintenance and killing. PD-1 thus contributes to the suppression of TIL function by inducing a state of impaired subcellular organization.
Collapse
Affiliation(s)
- Rachel Ambler
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Grace L Edmunds
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Sin Lih Tan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Silvia Cirillo
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Jane I Pernes
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Xiongtao Ruan
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jorge Huete-Carrasco
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Carissa C W Wong
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Jiahe Lu
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Juma Ward
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Giulia Toti
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Alan J Hedges
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Simon J Dovedi
- R&D Oncology, AstraZeneca, Granta Park, Cambridge CB21 6GH, UK
| | - Robert F Murphy
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA.,Departments of Biological Sciences, Biomedical Engineering and Machine Learning, Carnegie Mellon University, Pittsburgh, PA 15213, USA.,Freiburg Institute for Advanced Studies and Faculty of Biology, Albert Ludwig University of Freiburg, 79104 Freiburg, Germany
| | - David J Morgan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK.
| | - Christoph Wülfing
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
7
|
Mohammadzadeh A. Co-inhibitory receptors, transcription factors and tolerance. Int Immunopharmacol 2020; 84:106572. [DOI: 10.1016/j.intimp.2020.106572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 05/04/2020] [Indexed: 12/23/2022]
|
8
|
Kudo H, Tsuboi H, Asashima H, Takahashi H, Ono Y, Abe S, Honda F, Kondo Y, Wakasa Y, Takaiwa F, Takano M, Matsui M, Matsumoto I, Sumida T. Transgenic rice seeds expressing altered peptide ligands against the M3 muscarinic acetylcholine receptor suppress experimental sialadenitis-like Sjögren's syndrome. Mod Rheumatol 2019; 30:884-893. [PMID: 31490711 DOI: 10.1080/14397595.2019.1664368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Objective: We previously reported that Rag1-/- mice inoculated with splenocytes from M3 muscarinic acetylcholine receptor (M3R) knockout mice immunized with an M3R peptide mixture developed sialadenitis-like Sjögren's syndrome (M3R-induced sialadenitis [MIS]). We also found that intravenous administration of altered peptide ligand (APL) of N-terminal 1 (N1), which is one of the T-cell epitopes of M3R, suppressed MIS. In this study, we aimed to evaluate the suppressive ability and its mechanisms of rice seeds expressing N1-APL7 against MIS.Methods: Rice seeds expressing N1 and N1-APL7 were orally administered to MIS mice for 2 weeks. The changes in saliva flow and sialadenitis (salivary gland inflammation) were analyzed. The M3R-specific T-cell response in the spleen and the expression of regulatory molecules in the cervical lymph nodes and mesenteric lymph nodes were also analyzed.Results: Oral administration of N1-APL7-expressing rice seeds significantly recovered reduction in saliva flow and suppressed sialadenitis when compared with treatment with nontransgenic rice seeds and N1 rice seeds. IFNγ production from M3R-reactive T cells tended to decline in the N1-APL7 rice-treated group as compared with those in the other groups. In the N1-APL7 rice-treated group, the mRNA expression levels of Foxp3 in the cervical-lymph-node CD4+ T cells were higher than those in the other groups.Conclusion: Oral administration of N1-APL7-expressing rice suppressed MIS via suppression of M3R-specific IFNγ and IL-17 production and via enhancement of regulatory molecule expression.Key messagesWe generated N1-peptide- or N1-APL7-expressing rice seeds. Oral administration of N1-APL7-expressing rice seeds significantly recovered the reduction of saliva flow and suppressed sialadenitis via the suppression of M3R specific IFNγ and IL-17 production and via enhancement of regulatory T (Treg) cells.
Collapse
Affiliation(s)
- Hanae Kudo
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hiroto Tsuboi
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hiromitsu Asashima
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hiroyuki Takahashi
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yuko Ono
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Saori Abe
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Fumika Honda
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yuya Kondo
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yuhya Wakasa
- Plant Molecular Farming Unit, Division of Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Fumio Takaiwa
- Plant Molecular Farming Unit, Division of Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Makoto Takano
- Plant Molecular Farming Unit, Division of Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | | | - Isao Matsumoto
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
9
|
Hamuro L, Tirucherai GS, Crawford SM, Nayeem A, Pillutla RC, DeSilva BS, Leil TA, Thalhauser CJ. Evaluating a Multiscale Mechanistic Model of the Immune System to Predict Human Immunogenicity for a Biotherapeutic in Phase 1. AAPS JOURNAL 2019; 21:94. [PMID: 31342199 DOI: 10.1208/s12248-019-0361-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/28/2019] [Indexed: 02/06/2023]
Abstract
A mechanistic model of the immune response was evaluated for its ability to predict anti-drug antibody (ADA) and their impact on pharmacokinetics (PK) and pharmacodynamics (PD) for a biotherapeutic in a phase 1 clinical trial. Observed ADA incidence ranged from 33 to 67% after single doses and 27-50% after multiple doses. The model captured the single dose incidence well; however, there was overprediction after multiple dosing. The model was updated to include a T-regulatory (Treg) cell mediated tolerance, which reduced the overprediction (relative decrease in predicted incidence rate of 21.5-59.3% across multidose panels) without compromising the single dose predictions (relative decrease in predicted incidence rate of 0.6-13%). The Treg-adjusted model predicted no ADA impact on PK or PD, consistent with the observed data. A prospective phase 2 trial was simulated, including co-medication effects in the form of corticosteroid-induced immunosuppression. Predicted ADA incidences were 0-10%, depending on co-medication dosage. This work demonstrates the utility in applying an integrated, iterative modeling approach to predict ADA during different stages of clinical development.
Collapse
Affiliation(s)
- Lora Hamuro
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, 08543, USA
| | - Giridhar S Tirucherai
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, 08543, USA
| | - Sean M Crawford
- Bioanalytical Sciences, Translational Medicine, Bristol-Myers Squibb, Princeton, New Jersey, 08543, USA
| | - Akbar Nayeem
- Molecular Structure and Design, Bristol-Myers Squibb, Princeton, New Jersey, 08543, USA
| | - Renuka C Pillutla
- Bioanalytical Sciences, Translational Medicine, Bristol-Myers Squibb, Princeton, New Jersey, 08543, USA
| | - Binodh S DeSilva
- Analytical Strategy and Operations, Product Development, Bristol-Myers Squibb, Princeton, New Jersey, 08543, USA
| | - Tarek A Leil
- Quantitative Clinical Pharmacology, Bristol-Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey, 08543, USA
| | - Craig J Thalhauser
- Quantitative Clinical Pharmacology, Bristol-Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey, 08543, USA.
| |
Collapse
|
10
|
Okamura T, Yamamoto K, Fujio K. Early Growth Response Gene 2-Expressing CD4 +LAG3 + Regulatory T Cells: The Therapeutic Potential for Treating Autoimmune Diseases. Front Immunol 2018. [PMID: 29535721 PMCID: PMC5834469 DOI: 10.3389/fimmu.2018.00340] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Regulatory T cells (Tregs) are necessary for the maintenance of immune tolerance. Tregs are divided into two major populations: one is thymus derived and the other develops in the periphery. Among these Tregs, CD4+CD25+ Tregs, which mainly originate in the thymus, have been extensively studied. Transcription factor Foxp3 is well known as a master regulatory gene for the development and function of CD4+CD25+ Tregs. On the other hand, peripheral Tregs consist of distinct cell subsets including Foxp3-dependent extrathymically developed Tregs and interleukin (IL)-10-producing type I regulatory T (Tr1) cells. Lymphocyte activation gene 3 (LAG3) and CD49b are reliable cell surface markers for Tr1 cells. CD4+CD25−LAG3+ Tregs (LAG3+ Tregs) develop in the periphery and produce a large amount of IL-10. LAG3+ Tregs characteristically express the early growth response gene 2 (Egr2), a zinc-finger transcription factor, and exhibit its suppressive activity in a Foxp3-independent manner. Although Egr2 was known to be essential for hindbrain development and myelination of the peripheral nervous system, recent studies revealed that Egr2 plays vital roles in the induction of T cell anergy and also the suppressive activities of LAG3+ Tregs. Intriguingly, forced expression of Egr2 converts naive CD4+ T cells into IL-10-producing Tregs that highly express LAG3. Among the four Egr gene family members, Egr3 is thought to compensate for the function of Egr2. Recently, we reported that LAG3+ Tregs suppress humoral immune responses via transforming growth factor β3 production in an Egr2- and Egr3-dependent manner. In this review, we focus on the role of Egr2 in Tregs and also discuss its therapeutic potential for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan.,Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Omodho B, Miao T, Symonds ALJ, Singh R, Li S, Wang P. Transcription factors early growth response gene (Egr) 2 and 3 control inflammatory responses of tolerant T cells. IMMUNITY INFLAMMATION AND DISEASE 2018; 6:221-233. [PMID: 29314730 PMCID: PMC5946152 DOI: 10.1002/iid3.210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 01/07/2023]
Abstract
Introduction Impaired proliferation and production of IL2 are the hallmarks of experimental T cell tolerance. However, in most autoimmune diseases, auto‐reactive T cells do not display hyper proliferation, but inflammatory phenotypes. Methods We have now demonstrated that the transcription factors Egr2 and 3 are important for the control of inflammatory cytokine production by tolerant T cells, but not for tolerance induction. Results In the absence of Egr2 and 3, T cell tolerance, as measured by impaired proliferation and production of IL2, can still be induced, but tolerant T cells produced high levels of inflammatory cytokines. Egr2 and 3 regulate expression of differentiation repressors and directly inhibit T‐bet function in T cells. Indeed, decreased expression of differentiation repressors, such as Id3 and Tcf1, and increased expression of inflammatory transcription factors, such as RORγt and Bhlhe40 were found in Egr2/3 deficient T cells under tolerogenic conditions. In addition, T‐bet was co‐expressed with Egr2 in tolerant T cells and Egr2/3 defects leads to production of high levels of IFNγ in tolerant T cells. Conclusions Our findings demonstrated that despite impaired proliferation and IL2 production, tolerant T cells can display inflammatory responses in response to antigen stimulation and this is controlled at least partly by Egr2 and 3.
Collapse
Affiliation(s)
- Becky Omodho
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, UK.,Bioscience, Brunel University London, Kingston Lane, London, UK
| | - Tizong Miao
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, UK
| | - Alistair L J Symonds
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, UK
| | - Randeep Singh
- Bioscience, Brunel University London, Kingston Lane, London, UK
| | - Suling Li
- Bioscience, Brunel University London, Kingston Lane, London, UK
| | - Ping Wang
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, UK
| |
Collapse
|
12
|
Xiang L, Xin N, Yuan Y, Hou X, Chen J, Wei N, Gong P. Effect of follicular dendritic cell secreted protein on gene expression of human periodontal ligament cells. Arch Oral Biol 2017; 81:151-159. [PMID: 28544936 DOI: 10.1016/j.archoralbio.2017.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 04/27/2017] [Accepted: 05/14/2017] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The objective of this study was to investigate the specific roles of follicular dendritic cell secreted protein (FDC-SP), a protein exists in saliva, in the inhibition of calcium precipitation during periodontal regeneration, as well as affect phenotype expression of human periodontal ligament cells (hPDLCs) during the differentiation process. DESIGN To investigate this, we applied microarray technology to identify gene expression changes in hPDLCs transfected with FDC-SP and then clustered them according to their biological functions. RESULTS One hundred seventy-one genes were found differentially expressed by at least two-fold between FDC-SP -transfected and empty vector-transfected cells. Besides, genes encoding cell-cycle proteins, blood-related and cell differentiation-related proteins tended to be up-regulated after FDC-SP transfection, whereas cytokine/growth factors, signal transduction and metabolism-related genes tended to be down-regulated in hPDLCs overexpression FDC-SP. CONCLUSIONS The present study investigated FDC-SP's roles in hPDLCs' phenotype expression, via comparing the gene expression profiles between FDC-SP -transfected hPDLCs and empty vector-transfected cells upon microarray analysis. hPDLCs overexpression FDC-SP appear to display different gene expression patterns. In all, these observations showed a potential of FDC-SP in the maintenance of PDL homeostasis and its ultimate contribution to periodontal would-healing processes.
Collapse
Affiliation(s)
- Lin Xiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Na Xin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Ying Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xiaogang Hou
- College of Hydraulic and Hydroelectric Engineering, Sichuan University, Chengdu 610041, China.
| | - Junwei Chen
- Department of Energy and Resources Engineering, College of Engineering, Peking University, Beijing 100000, China.
| | - Na Wei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Ping Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
13
|
Miao T, Symonds ALJ, Singh R, Symonds JD, Ogbe A, Omodho B, Zhu B, Li S, Wang P. Egr2 and 3 control adaptive immune responses by temporally uncoupling expansion from T cell differentiation. J Exp Med 2017; 214:1787-1808. [PMID: 28487311 PMCID: PMC5460991 DOI: 10.1084/jem.20160553] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 11/23/2016] [Accepted: 03/24/2017] [Indexed: 12/30/2022] Open
Abstract
Miao et al. report a checkpoint mediated by Egr2 and 3 that controls the transition between T cell clonal expansion and differentiation by regulating genes involved in proliferation and differentiation, which is essential for optimal immune responses with limited immunopathology. Egr2 and 3 are important for maintaining immune homeostasis. Here we define a fundamental function of Egr2 and 3 operating as a checkpoint that controls the transition between clonal expansion and differentiation of effector T cells. Egr2 and 3 deficiency resulted in defective clonal expansion but hyperactivation and excessive differentiation of T cells in response to viral infection. Conversely, sustained Egr2 expression enhanced expansion but severely impaired effector differentiation. Egr2 bound to and controlled the expression of genes regulating proliferation (Myc and Myb) and differentiation repressors (Bcl6, Id3), while repressing transcription factors required for effector function (Zeb2, RORa, RORc, and Bhlhe40). Egr2 and 3 expression in T cells was regulated reciprocally by antigen and IFNγ, providing a mechanism for adjusting proliferation and differentiation of individual T cells. Thus, Egr2 and 3 are upstream regulators of effector CD4 and CD8 T cells that are essential for optimal responses with limited immunopathology.
Collapse
Affiliation(s)
- Tizong Miao
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK
| | - Alistair L J Symonds
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK
| | - Randeep Singh
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK.,Bioscience, Brunel University, Uxbridge UB8 3PH, England, UK
| | - Janine D Symonds
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology (CoMPLEX), University College London, London WC1E 6BT, England, UK
| | - Ane Ogbe
- Bioscience, Brunel University, Uxbridge UB8 3PH, England, UK
| | - Becky Omodho
- Bioscience, Brunel University, Uxbridge UB8 3PH, England, UK
| | - Bo Zhu
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK.,Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Suling Li
- Bioscience, Brunel University, Uxbridge UB8 3PH, England, UK
| | - Ping Wang
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK
| |
Collapse
|
14
|
Singh R, Miao T, Symonds ALJ, Omodho B, Li S, Wang P. Egr2 and 3 Inhibit T-bet-Mediated IFN-γ Production in T Cells. THE JOURNAL OF IMMUNOLOGY 2017; 198:4394-4402. [PMID: 28455436 PMCID: PMC5439026 DOI: 10.4049/jimmunol.1602010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/26/2017] [Indexed: 12/31/2022]
Abstract
T-bet is important for differentiation of cytotoxic CD8 and Th1 CD4 T cells. We have discovered that Egr2 and 3 are potent inhibitors of T-bet function in CD4 and CD8 effector T cells. Egr2 and 3 were essential to suppress Th1 differentiation in Th2 and Th17 conditions in vitro and also to control IFN-γ–producing CD4 and CD8 T cells in response to virus infection. Together with Egr2 and 3, T-bet is induced in naive T cells by Ag stimulation, but Egr2 and 3 expression was inhibited by Th1–inducing cytokines. We found that Egr2 and 3 physically interact with the T-box domain of T-bet, blocking T-bet DNA binding and inhibiting T-bet–mediated production of IFN-γ. Thus, Egr2 and 3 are antagonists of T-bet function in effector T cells and are important for the control of inflammatory responses of T cells.
Collapse
Affiliation(s)
- Randeep Singh
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom; and.,Bioscience, Brunel University London, London UB8 3PH, United Kingdom
| | - Tizong Miao
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom; and
| | - Alistair L J Symonds
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom; and
| | - Becky Omodho
- Bioscience, Brunel University London, London UB8 3PH, United Kingdom
| | - Suling Li
- Bioscience, Brunel University London, London UB8 3PH, United Kingdom
| | - Ping Wang
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom; and
| |
Collapse
|
15
|
Egr2 and Egr3 in regulatory T cells cooperatively control systemic autoimmunity through Ltbp3-mediated TGF-β3 production. Proc Natl Acad Sci U S A 2016; 113:E8131-E8140. [PMID: 27911796 DOI: 10.1073/pnas.1611286114] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease characterized by multiorgan inflammation induced by autoantibodies. Early growth response gene 2 (Egr2), a transcription factor essential for T-cell anergy induction, controls systemic autoimmunity in mice and humans. We have previously identified a subpopulation of CD4+ regulatory T cells, CD4+CD25-LAG3+ cells, that characteristically express both Egr2 and LAG3 and control mice model of lupus via TGF-β3 production. However, due to the mild phenotype of lymphocyte-specific Egr2-deficient mice, the presence of an additional regulator has been speculated. Here, we show that Egr2 and Egr3 expressed in T cells cooperatively prevent humoral immune responses by supporting TGF-β3 secretion. T cell-specific Egr2/Egr3 double-deficient (Egr2/3DKO) mice spontaneously developed an early onset lupus-like disease that was more severe than in T cell-specific Egr2-deficient mice. In accordance with the observation that CD4+CD25-LAG3+ cells from Egr2/3DKO mice completely lost the capacity to produce TGF-β3, the excessive germinal center reaction in Egr2/3DKO mice was suppressed by the adoptive transfer of WT CD4+CD25-LAG3+ cells or treatment with a TGF-β3-expressing vector. Intriguingly, latent TGF-β binding protein (Ltbp)3 expression maintained by Egr2 and Egr3 was required for TGF-β3 production from CD4+CD25-LAG3+ cells. Because Egr2 and Egr3 did not demonstrate cell intrinsic suppression of the development of follicular helper T cells, Egr2- and Egr3-dependent TGF-β3 production by CD4+CD25-LAG3+ cells is critical for controlling excessive B-cell responses. The unique attributes of Egr2/Egr3 in T cells may provide an opportunity for developing novel therapeutics for autoantibody-mediated diseases including SLE.
Collapse
|
16
|
Candia M, Kratzer B, Pickl WF. On Peptides and Altered Peptide Ligands: From Origin, Mode of Action and Design to Clinical Application (Immunotherapy). Int Arch Allergy Immunol 2016; 170:211-233. [PMID: 27642756 PMCID: PMC7058415 DOI: 10.1159/000448756] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
T lymphocytes equipped with clonotypic T cell antigen receptors (TCR) recognize immunogenic peptides only when presented in the context of their own major histocompatibility complex (MHC) molecules. Peptide loading to MHC molecules occurs in intracellular compartments (ER for class I and MIIC for class II molecules) and relies on the interaction of the respective peptides and peptide binding pockets on MHC molecules. Those peptide residues not engaged in MHC binding point towards the TCR screening for possible peptide MHC complex binding partners. Natural or intentional modification of both MHC binding registers and TCR interacting residues of peptides - leading to the formation of altered peptide ligands (APLs) - might alter the way peptides interact with TCRs and hence influence subsequent T cell activation events, and consequently T cell effector functions. This review article summarizes how APLs were detected and first described, current concepts of how APLs modify T cellular signaling, which biological mechanisms might force the generation of APLs in vivo, and how peptides and APLs might be used for the benefit of patients suffering from allergic or autoimmune diseases.
Collapse
Affiliation(s)
- Martín Candia
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Immunomodulation, Vienna, Austria
| |
Collapse
|
17
|
White AM, Wraith DC. Tr1-Like T Cells - An Enigmatic Regulatory T Cell Lineage. Front Immunol 2016; 7:355. [PMID: 27683580 PMCID: PMC5021682 DOI: 10.3389/fimmu.2016.00355] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/31/2016] [Indexed: 11/30/2022] Open
Abstract
The immune system evolved to respond to foreign invaders and prevent autoimmunity to self-antigens. Several types of regulatory T cells facilitate the latter process. These include a subset of Foxp3− CD4+ T cells able to secrete IL-10 in an antigen-specific manner, type 1 regulatory (Tr1) T cells. Although their suppressive function has been confirmed both in vitro and in vivo, their phenotype remains poorly defined. It has been suggested that the surface markers LAG-3 and CD49b are biomarkers for murine and human Tr1 cells. Here, we discuss these findings in the context of our data regarding the expression pattern of inhibitory receptors (IRs) CD49b, TIM-3, PD-1, TIGIT, LAG-3, and ICOS on Tr1-like human T cells generated in vitro from CD4+ memory T cells stimulated with αCD3 and αCD28 antibodies. We found that there were no differences in IR expression between IL-10+ and IL-10− T cells. However, CD4+IL-10+ T cells isolated ex vivo, following a short stimulation and cytokine secretion assay, contained significantly higher proportions of TIM-3+ and PD-1+ cells. They also expressed significantly higher TIGIT mRNA and showed a trend toward increased TIM-3 mRNA levels. These data led us to conclude that large pools of IRs may be stored intracellularly; hence, they may not represent ideal candidates as cell surface biomarkers for Tr1-like T cells.
Collapse
Affiliation(s)
| | - David C Wraith
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK; Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| |
Collapse
|
18
|
Kozela E, Juknat A, Gao F, Kaushansky N, Coppola G, Vogel Z. Pathways and gene networks mediating the regulatory effects of cannabidiol, a nonpsychoactive cannabinoid, in autoimmune T cells. J Neuroinflammation 2016; 13:136. [PMID: 27256343 PMCID: PMC4891926 DOI: 10.1186/s12974-016-0603-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 05/27/2016] [Indexed: 11/29/2022] Open
Abstract
Background Our previous studies showed that the non-psychoactive cannabinoid, cannabidiol (CBD), ameliorates the clinical symptoms in mouse myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis model of multiple sclerosis (MS) as well as decreases the memory MOG35-55-specific T cell (TMOG) proliferation and cytokine secretion including IL-17, a key autoimmune factor. The mechanisms of these activities are currently poorly understood. Methods Herein, using microarray-based gene expression profiling, we describe gene networks and intracellular pathways involved in CBD-induced suppression of these activated memory TMOG cells. Encephalitogenic TMOG cells were stimulated with MOG35-55 in the presence of spleen-derived antigen presenting cells (APC) with or without CBD. mRNA of purified TMOG was then subjected to Illumina microarray analysis followed by ingenuity pathway analysis (IPA), weighted gene co-expression network analysis (WGCNA) and gene ontology (GO) elucidation of gene interactions. Results were validated using qPCR and ELISA assays. Results Gene profiling showed that the CBD treatment suppresses the transcription of a large number of proinflammatory genes in activated TMOG. These include cytokines (Xcl1, Il3, Il12a, Il1b), cytokine receptors (Cxcr1, Ifngr1), transcription factors (Ier3, Atf3, Nr4a3, Crem), and TNF superfamily signaling molecules (Tnfsf11, Tnfsf14, Tnfrsf9, Tnfrsf18). “IL-17 differentiation” and “IL-6 and IL-10-signaling” were identified among the top processes affected by CBD. CBD increases a number of IFN-dependent transcripts (Rgs16, Mx2, Rsad2, Irf4, Ifit2, Ephx1, Ets2) known to execute anti-proliferative activities in T cells. Interestingly, certain MOG35-55 up-regulated transcripts were maintained at high levels in the presence of CBD, including transcription factors (Egr2, Egr1, Tbx21), cytokines (Csf2, Tnf, Ifng), and chemokines (Ccl3, Ccl4, Cxcl10) suggesting that CBD may promote exhaustion of memory TMOG cells. In addition, CBD enhanced the transcription of T cell co-inhibitory molecules (Btla, Lag3, Trat1, and CD69) known to interfere with T/APC interactions. Furthermore, CBD enhanced the transcription of oxidative stress modulators with potent anti-inflammatory activity that are controlled by Nfe2l2/Nrf2 (Mt1, Mt2a, Slc30a1, Hmox1). Conclusions Microarray-based gene expression profiling demonstrated that CBD exerts its immunoregulatory effects in activated memory TMOG cells via (a) suppressing proinflammatory Th17-related transcription, (b) by promoting T cell exhaustion/tolerance, (c) enhancing IFN-dependent anti-proliferative program, (d) hampering antigen presentation, and (d) inducing antioxidant milieu resolving inflammation. These findings put forward mechanism by which CBD exerts its anti-inflammatory effects as well as explain the beneficial role of CBD in pathological memory T cells and in autoimmune diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0603-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ewa Kozela
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel. .,Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel.
| | - Ana Juknat
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel.,Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Fuying Gao
- Departments of Psychiatry and Neurology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Nathali Kaushansky
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Giovanni Coppola
- Departments of Psychiatry and Neurology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Zvi Vogel
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel.,Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
19
|
Northrup L, Christopher MA, Sullivan BP, Berkland C. Combining antigen and immunomodulators: Emerging trends in antigen-specific immunotherapy for autoimmunity. Adv Drug Deliv Rev 2016; 98:86-98. [PMID: 26546466 DOI: 10.1016/j.addr.2015.10.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/05/2023]
Abstract
A majority of current therapies for autoimmune diseases are general immunosuppressants, which can compromise patient response to opportunistic infection and lead to adverse events. Using antigen-specific immunotherapy (ASIT) to selectively disarm autoimmune diseases, without suppressing the global immune response, would be a transformative therapy for patients. ASIT has been used historically in allergy hyposensitization therapy to induce tolerance to an allergen. Similar strategies to induce immune tolerance toward autoantigens responsible for autoimmune disease have been attempted but have yielded limited clinical success. Recent studies of ASIT for autoimmunity have explored combination therapy, combining the disease-causing autoantigen with an immunomodulatory compound. ASIT combination therapy may direct the immune response in an antigen-specific manner, potentially reversing the root cause of autoimmunity while limiting side effects. This review analyzes recent advances in ASIT applied to autoimmune diseases, emphasizing current combination therapies and future strategies.
Collapse
Affiliation(s)
- Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Matthew A Christopher
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Bradley P Sullivan
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
20
|
Malek Abrahimians E, Carlier VA, Vander Elst L, Saint-Remy JMR. MHC Class II-Restricted Epitopes Containing an Oxidoreductase Activity Prompt CD4(+) T Cells with Apoptosis-Inducing Properties. Front Immunol 2015; 6:449. [PMID: 26388872 PMCID: PMC4556975 DOI: 10.3389/fimmu.2015.00449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/18/2015] [Indexed: 12/21/2022] Open
Abstract
Abrogating an unwanted immune response toward a specific antigen without compromising the entire immune system is a hoped-for goal in immunotherapy. Instead of manipulating dendritic cells and suppressive regulatory T cells, depleting effector T cells or blocking their co-stimulatory pathways, we describe a method to specifically inhibit the presentation of an antigen eliciting an unwanted immune reaction. Inclusion of an oxidoreductase motif within the flanking residues of MHC class II epitopes polarizes CD4(+) T cells to cytolytic cells capable of inducing apoptosis in antigen presenting cells (APCs) displaying cognate peptides through MHC class II molecules. This novel function results from an increased synapse formation between both cells. Moreover, these cells eliminate by apoptosis bystander CD4(+) T cells activated at the surface of the APC. We hypothesize that they would thereby block the recruitment of cells of alternative specificity for the same autoantigen or cells specific for another antigen associated with the pathology, providing a system by which response against multiple antigens linked with the same disease can be suppressed. These findings open the way toward a novel form of antigen-specific immunosuppression.
Collapse
Affiliation(s)
- Elin Malek Abrahimians
- Center for Molecular and Vascular Biology, University of Leuven , Leuven , Belgium ; ImCyse SA , Leuven , Belgium
| | - Vincent A Carlier
- Center for Molecular and Vascular Biology, University of Leuven , Leuven , Belgium ; ImCyse SA , Leuven , Belgium
| | - Luc Vander Elst
- Center for Molecular and Vascular Biology, University of Leuven , Leuven , Belgium ; ImCyse SA , Leuven , Belgium
| | - Jean-Marie R Saint-Remy
- Center for Molecular and Vascular Biology, University of Leuven , Leuven , Belgium ; ImCyse SA , Leuven , Belgium
| |
Collapse
|
21
|
Kozela E, Juknat A, Kaushansky N, Ben-Nun A, Coppola G, Vogel Z. Cannabidiol, a non-psychoactive cannabinoid, leads to EGR2-dependent anergy in activated encephalitogenic T cells. J Neuroinflammation 2015; 12:52. [PMID: 25880134 PMCID: PMC4363052 DOI: 10.1186/s12974-015-0273-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/26/2015] [Indexed: 11/16/2022] Open
Abstract
Background Cannabidiol (CBD), the main non-psychoactive cannabinoid, has been previously shown by us to ameliorate clinical symptoms and to decrease inflammation in myelin oligodendrocyte glycoprotein (MOG)35-55-induced mouse experimental autoimmune encephalomyelitis model of multiple sclerosis as well as to decrease MOG35-55-induced T cell proliferation and IL-17 secretion. However, the mechanisms of CBD anti-inflammatory activities are unclear. Methods Here we analyzed the effects of CBD on splenocytes (source of accessory T cells and antigen presenting cells (APC)) co-cultured with MOG35-55-specific T cells (TMOG) and stimulated with MOG35-55. Using flow cytometry, we evaluated the expression of surface activation markers and inhibitory molecules on T cells and B cells. TMOG cells were purified using CD4 positive microbead selection and submitted for quantitative PCR and microarray of mRNA transcript analyzes. Cell signaling studies in purified TMOG were carried out using immunoblotting. Results We found that CBD leads to upregulation of CD69 and lymphocyte-activation gene 3 (LAG3) regulatory molecules on CD4+CD25− accessory T cells. This subtype of CD4+CD25−CD69+LAG3+ T cells has been recognized as induced regulatory phenotype promoting anergy in activated T cells. Indeed, we observed that CBD treatment results in upregulation of EGR2 (a key T cell anergy inducer) mRNA transcription in stimulated TMOG cells. This was accompanied by elevated levels of anergy promoting genes such as IL-10 (anti-inflammatory cytokine), STAT5 (regulatory factor), and LAG3 mRNAs, as well as of several enhancers of cell cycle arrest (such as Nfatc1, Casp4, Cdkn1a, and Icos). Moreover, CBD exposure leads to a decrease in STAT3 and to an increase in STAT5 phosphorylation in TMOG cells, positive and negative regulators of Th17 activity, respectively. In parallel, we observed decreased levels of major histocompatibility complex class II (MHCII), CD25, and CD69 on CD19+ B cells following CBD treatment, showing diminished antigen presenting capabilities of B cells and reduction in their pro-inflammatory functions. Conclusions Our data suggests that CBD exerts its immunoregulatory effects via induction of CD4+CD25−CD69+LAG3+ cells in MOG35-55-activated APC/TMOG co-cultures. This is accompanied by EGR2-dependent anergy of stimulated TMOG cells as well as a switch in their intracellular STAT3/STAT5 activation balance leading to the previously observed decrease in Th17 activity.
Collapse
Affiliation(s)
- Ewa Kozela
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Ana Juknat
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Nathali Kaushansky
- Neurobiology Department, Weizmann Institute of Science, Rehovot, Israel.
| | - Avraham Ben-Nun
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.
| | | | - Zvi Vogel
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Neurobiology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
22
|
Pletinckx K, Vaeth M, Schneider T, Beyersdorf N, Hünig T, Berberich-Siebelt F, Lutz MB. Immature dendritic cells convert anergic nonregulatory T cells into Foxp3- IL-10+ regulatory T cells by engaging CD28 and CTLA-4. Eur J Immunol 2014; 45:480-91. [PMID: 25382658 DOI: 10.1002/eji.201444991] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/29/2014] [Accepted: 11/06/2014] [Indexed: 12/17/2022]
Abstract
Anergic T cells can survive for long time periods passively in a hyporesponsive state without obvious active functions. Thus, the immunological reason for their maintenance is unclear. Here, we induced peptide-specific anergy in T cells from mice by coculturing these cells with immature murine dendritic cells (DCs). We found that these anergic, nonsuppressive IL-10(-) Foxp3(-) CTLA-4(+) CD25(low) Egr2(+) T cells could be converted into suppressive IL-10(+) Foxp3(-) CTLA-4(+) CD25(high) Egr2(+) cells resembling type-1 Treg cells (Tr1) when stimulated a second time by immature DCs in vitro. Addition of TGF-β during anergy induction favored Foxp3(+) Treg-cell induction, while TGF-β had little effect when added to the second stimulation. Expression of both CD28 and CTLA-4 molecules on anergic T cells was required to allow their conversion into Tr1-like cells. Suppressor activity was enabled via CD28-mediated CD25 upregulation, acting as an IL-2 sink, together with a CTLA-4-mediated inhibition of NFATc1/α activation to shut down IL-2-mediated proliferation. Together, these data provide evidence and mechanistical insights into how persistent anergic T cells may serve as a resting memory pool for Tr1-like cells.
Collapse
Affiliation(s)
- Katrien Pletinckx
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Burton BR, Britton GJ, Fang H, Verhagen J, Smithers B, Sabatos-Peyton CA, Carney LJ, Gough J, Strobel S, Wraith DC. Sequential transcriptional changes dictate safe and effective antigen-specific immunotherapy. Nat Commun 2014; 5:4741. [PMID: 25182274 PMCID: PMC4167604 DOI: 10.1038/ncomms5741] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 07/18/2014] [Indexed: 01/13/2023] Open
Abstract
Antigen-specific immunotherapy combats autoimmunity or allergy by reinstating immunological tolerance to target antigens without compromising immune function. Optimization of dosing strategy is critical for effective modulation of pathogenic CD4+ T-cell activity. Here we report that dose escalation is imperative for safe, subcutaneous delivery of the high self-antigen doses required for effective tolerance induction and elicits anergic, interleukin (IL)-10-secreting regulatory CD4+ T cells. Analysis of the CD4+ T-cell transcriptome, at consecutive stages of escalating dose immunotherapy, reveals progressive suppression of transcripts positively regulating inflammatory effector function and repression of cell cycle pathways. We identify transcription factors, c-Maf and NFIL3, and negative co-stimulatory molecules, LAG-3, TIGIT, PD-1 and TIM-3, which characterize this regulatory CD4+ T-cell population and whose expression correlates with the immunoregulatory cytokine IL-10. These results provide a rationale for dose escalation in T-cell-directed immunotherapy and reveal novel immunological and transcriptional signatures as surrogate markers of successful immunotherapy. Dose escalation in antigen-specific therapies is recognized as safe and effective, but the underlying effects of dosing variables on the immune system are not understood. Here, the authors demonstrate that dose escalation causes sequential modulation of gene expression among antigen-specific lymphocytes.
Collapse
Affiliation(s)
- Bronwen R Burton
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Graham J Britton
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Hai Fang
- Computational Genomics Group, Department of Computer Science, University of Bristol, Bristol BS8 1UB, UK
| | - Johan Verhagen
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Ben Smithers
- Computational Genomics Group, Department of Computer Science, University of Bristol, Bristol BS8 1UB, UK
| | | | - Laura J Carney
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Julian Gough
- Computational Genomics Group, Department of Computer Science, University of Bristol, Bristol BS8 1UB, UK
| | - Stephan Strobel
- Division of Biomedical Sciences, Institute of Child Health, University College London, London WC1N 1EH, UK
| | - David C Wraith
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
24
|
Abstract
Anergy is a long-term stable state of T-lymphocyte unresponsiveness to antigenic stimulation associated with the blockade of IL-2 production and proliferation. Anergy is a pathway of peripheral tolerance formation. In this review, mechanisms underlying T-cell tolerization are considered in a classical in vitro model of clonal anergy, and these mechanisms are compared with different pathways of anergy induction in vivo. Special attention is given to regulatory T-lymphocytes because, on one hand, anergy is a specific feature of these cells, and on the other hand anergy is also a mechanism of their action on target cells - effector T-lymphocytes. The role of this phenomenon in the differentiation of regulatory T-cells and also in the development of activation-induced apoptosis in effector T-lymphocytes is discussed.
Collapse
Affiliation(s)
- E M Kuklina
- Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, 614081 Perm, Russia.
| |
Collapse
|
25
|
Iwasaki Y, Fujio K, Okamura T, Yanai A, Sumitomo S, Shoda H, Tamura T, Yoshida H, Charnay P, Yamamoto K. Egr-2 transcription factor is required for Blimp-1-mediated IL-10 production in IL-27-stimulated CD4+ T cells. Eur J Immunol 2013; 43:1063-73. [PMID: 23349024 DOI: 10.1002/eji.201242942] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 01/14/2013] [Accepted: 01/22/2013] [Indexed: 11/09/2022]
Abstract
Interleukin-27 (IL-27) suppresses immune responses through inhibition of the development of IL-17 producing Th17 cells and induction of IL-10 production. We previously showed that forced expression of early growth response gene 2 (Egr-2), a transcription factor required for T-cell anergy induction, induces IL-10 and lymphocyte activation gene 3 expression and confers regulatory activity on CD4(+) T cells in vivo. Here, we evaluated the role of Egr-2 in IL-27-induced IL-10 production. Among various IL-10-inducing factors, only IL-27 induced high levels of Egr-2 and lymphocyte activation gene 3 expression. Intriguingly, IL-27 failed to induce IL-10 in Egr-2-deficient T cells. IL-27-mediated induction of Prdm1 that codes B lymphocyte induced maturation protein-1, a transcriptional regulator important for IL-10 production in CD4(+) T cells, was also impaired in the absence of Egr-2. Although IL-27-mediated IL-10 induction was dependent on both STAT1 and STAT3, only STAT3 was required for IL-27-mediated Egr-2 induction. These results suggest that IL-27 signal transduction through Egr-2 and B lymphocyte induced maturation protein-1 plays an important role in IL-10 production. Furthermore, Egr-2-deficient CD4(+) T cells showed dysregulated production of IFN-γ and IL-17 in response to IL-27 stimulation. Therefore, Egr-2 may play key roles in controlling the balance between regulatory and effector cytokines.
Collapse
Affiliation(s)
- Yukiko Iwasaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Rzepecka J, Siebeke I, Coltherd JC, Kean DE, Steiger CN, Al-Riyami L, McSharry C, Harnett MM, Harnett W. The helminth product, ES-62, protects against airway inflammation by resetting the Th cell phenotype. Int J Parasitol 2013; 43:211-23. [PMID: 23291461 PMCID: PMC3584281 DOI: 10.1016/j.ijpara.2012.12.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 12/01/2012] [Accepted: 12/03/2012] [Indexed: 12/23/2022]
Abstract
We previously demonstrated inhibition of ovalbumin-induced allergic airway hyper-responsiveness in the mouse using ES-62, a phosphorylcholine-containing glycoprotein secreted by the filarial nematode, Acanthocheilonema viteae. This inhibition correlated with ES-62-induced mast cell desensitisation, although the degree to which this reflected direct targeting of mast cells remained unclear as suppression of the Th2 phenotype of the inflammatory response, as measured by eosinophilia and IL-4 levels in the lungs, was also observed. We now show that inhibition of the lung Th2 phenotype is reflected in ex vivo analyses of draining lymph node recall cultures and accompanied by a decrease in the serum levels of total and ovalbumin-specific IgE. Moreover, ES-62 also suppresses the lung infiltration by neutrophils that is associated with severe asthma and is generally refractory to conventional anti-inflammatory therapies, including steroids. Protection against Th2-associated airway inflammation does not reflect induction of regulatory T cell responses (there is no increased IL-10 or Foxp3 expression) but rather a switch in polarisation towards increased Tbet expression and IFNγ production. This ES-62-driven switch in the Th1/Th2 balance is accompanied by decreased IL-17 responses, a finding in line with reports that IFNγ and IL-17 are counter-regulatory. Consistent with ES-62 mediating its effects via IFNγ-mediated suppression of pathogenic Th2/Th17 responses, we found that neutralising anti-IFNγ antibodies blocked protection against airway inflammation in terms of pro-inflammatory cell infiltration, particularly by neutrophils, and lung pathology. Collectively, these studies indicate that ES-62, or more likely small molecule analogues, could have therapeutic potential in asthma, in particular for those subtypes of patients (e.g. smokers, steroid-resistant) who are refractory to current treatments.
Collapse
Affiliation(s)
- Justyna Rzepecka
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Miao T, Raymond M, Bhullar P, Ghaffari E, Symonds ALJ, Meier UC, Giovannoni G, Li S, Wang P. Early growth response gene-2 controls IL-17 expression and Th17 differentiation by negatively regulating Batf. THE JOURNAL OF IMMUNOLOGY 2012. [PMID: 23203924 DOI: 10.4049/jimmunol.1200868] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Early growth response gene (Egr)-2 is important for the maintenance of T cell homeostasis and controls the development of autoimmune disease. However, the underlying mechanisms are unknown. We have now discovered that Egr-2, which is induced by TGF-β and IL-6, negatively regulates the expression of IL-17, but not IL-2 or IFN-γ, in effector T cells. In the absence of Egr-2, CD4 T cells produce high levels of Th17 cytokines, which renders mice susceptible to experimental autoimmune encephalomyelitis induction. T cells lacking Egr-2 show increased propensity for Th17, but not Th1 or Th2, differentiation. Control of IL-17 expression and Th17 differentiation by Egr-2 is due to inhibition of Batf, a transcription factor that regulates IL-17 expression and Th17 differentiation. Egr-2 interacts with Batf in CD4 T cells and suppresses its interaction with DNA sequences derived from the IL-17 promoter, whereas the activation of STAT3 and expression of retinoic acid-related orphan receptor γt are unchanged in Th17 cells in the absence of Egr-2. Thus, Egr-2 plays an important role to intrinsically control Th17 differentiation. We also found that CD4 T cells from multiple sclerosis patients have reduced expression of Egr-2 and increased expression of IL-17 following stimulation with anti-CD3 in vitro. Collectively, our results demonstrate that Egr-2 is an intrinsic regulator that controls Th17 differentiation by inhibiting Batf activation, which may be important for the control of multiple sclerosis development.
Collapse
Affiliation(s)
- Tizong Miao
- Blizard Institute of Cell and Molecular Science, Barts and London School of Medicine and Dentistry, London E1 2AD, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Li S, Miao T, Sebastian M, Bhullar P, Ghaffari E, Liu M, Symonds ALJ, Wang P. The transcription factors Egr2 and Egr3 are essential for the control of inflammation and antigen-induced proliferation of B and T cells. Immunity 2012; 37:685-96. [PMID: 23021953 PMCID: PMC3477314 DOI: 10.1016/j.immuni.2012.08.001] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 05/21/2012] [Accepted: 08/02/2012] [Indexed: 12/19/2022]
Abstract
Lymphocytes provide optimal responses against pathogens with minimal inflammatory pathology. However, the intrinsic mechanisms regulating these responses are unknown. Here, we report that deletion of both transcription factors Egr2 and Egr3 in lymphocytes resulted in a lethal autoimmune syndrome with excessive serum proinflammatory cytokines but also impaired antigen receptor-induced proliferation of B and T cells. Egr2- and Egr3-defective B and T cells had hyperactive signal transducer and activator of transcription-1 (STAT1) and STAT3 while antigen receptor-induced activation of transcription factor AP-1 was severely impaired. We discovered that Egr2 and/or Egr3 directly induced expression of suppressor of cytokine signaling-1 (SOCS1) and SOCS3, inhibitors of STAT1 and STAT3, and also blocked the function of Batf, an AP-1 inhibitor, in B and T cells. Thus, Egr2 and Egr3 regulate B and T cell function in adaptive immune responses and homeostasis by promoting antigen receptor signaling and controlling inflammation.
Collapse
|
29
|
Liu M, Miao T, Zhu H, Symonds ALJ, Li L, Schurich A, Maini MK, Zhang J, Kennedy PTF, Li S, Wang P. IL-2-engineered nano-APC effectively activates viral antigen-mediated T cell responses from chronic hepatitis B virus-infected patients. THE JOURNAL OF IMMUNOLOGY 2011; 188:1534-43. [PMID: 22210908 DOI: 10.4049/jimmunol.1102709] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Impaired function of virus-specific T cells resulting from virus persistence is one of the major mechanisms underlying the development of chronic hepatitis B viral infection. Previously, we found that IL-2 can restore the effector function of T cells rendered tolerant by Ag persistence. However, systemic administration of IL-2 induces organ pathology and expansion of T regulatory cells. In this study, we show that nano-APC with engineered HLA alleles and IL-2 deliver peptide-MHC complexes, costimulatory molecules, and IL-2 to Ag-responding T cells, resulting in enhanced expression of CD25 and activation of TCR signaling pathways, while suppressing PD-1 expression on viral-responding CD8 T cells from chronic hepatitis B virus patients. The enhanced activation of CD4 and CD8 T cells induced by IL-2-nano-APC was Ag dependent and IL-2-nano-APC did not affect T regulatory cells. At a size of 500 nm, the nano-APC effectively induce immune synapse formation on Ag-specific T cells and accumulate as free particles in the lymphoid organs. These attributes of IL-2-nano-APC or other bioadjuvant-engineered nano-APC have profound implications for their use as a therapeutic strategy in the treatment of chronic hepatitis B virus infection or other chronic viral diseases.
Collapse
Affiliation(s)
- Mengya Liu
- Division of Bioscience, Brunel University, London UB8 3PH, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Zeiser R, Penack O, Holler E, Idzko M. Danger signals activating innate immunity in graft-versus-host disease. J Mol Med (Berl) 2011; 89:833-45. [DOI: 10.1007/s00109-011-0767-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/29/2011] [Accepted: 05/03/2011] [Indexed: 12/20/2022]
|
31
|
Dominguez MDC, Lorenzo N, Barbera A, Darrasse-Jeze G, Hernández MV, Torres A, Hernández I, Gil R, Klatzmann D, Padrón G. An altered peptide ligand corresponding to a novel epitope from heat-shock protein 60 induces regulatory T cells and suppresses pathogenic response in an animal model of adjuvant-induced arthritis. Autoimmunity 2011; 44:471-82. [PMID: 21370936 DOI: 10.3109/08916934.2010.550590] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Induction of immune tolerance as therapeutic approach for autoimmune diseases constitutes a current research focal point. In this sense, we aimed to evaluate an altered peptide ligand (APL) for induction of peripheral tolerance in patients with rheumatoid arthritis (RA). A novel T-cell epitope from human heat-shock protein 60 (Hsp60), an autoantigen involved in the pathogenesis of RA, was identified by bioinformatics tools and an APL was design starting from this epitope. We investigated the ability of this APL for inducing regulatory T cells (Treg cells) in mice and evaluated the therapeutic effect of this peptide in an adjuvant-induced arthritis (AA) rat model. Clinical score, TNFα levels and histopathology were monitored, as well as the capacity of this APL for inducing Treg cells. Finally, the potentialities of the APL for inducing Treg cells were evaluated in ex vivo assays using mononuclear cells isolated from peripheral blood (PBMC). The APL induced an increase of the proportions of Treg cells in the draining lymph nodes of the injected site in mice. The APL efficiently inhibited the course of AA, with significant reduction of the clinical and histopathology score. This effect was associated with an increase of the proportions of Treg cells and a decrease of TNFα levels in spleen. Finally, stimulation of PBMCs from RA patients by the APL increases the proportions of the CD4(+)CD25(high)FoxP3(+) Treg cells. These results indicate a therapeutic potentiality of APL and support further investigation of this candidate drug for treatment of RA.
Collapse
|
32
|
Abstract
The development of Th1 lymphocytes is essential for cell-mediated immunity and resistance against intracellular pathogens. However, if left unregulated, the same response can cause serious damage to host tissues and lead to mortality. A number of different paracrine regulatory mechanisms involving distinct myeloid and lymphoid subpopulations have been implicated in controlling excessive secretion of inflammatory cytokines by Th1 cells. Much of this work has focused on interleukin (IL)-10, a cytokine with broad anti-inflammatory properties, one of which is to counteract the function of Th1 lymphocytes. While studying the role of IL-10 in regulating immunopathology during infection with the intracellular parasite Toxoplasma gondii, we discovered that the host-protective IL-10 derives in an autocrine manner from conventional interferon-gamma (IFN-gamma)-producing T-bet(+) Foxp3(neg) Th1 cells. In the following review, we will discuss these findings that support the general concept that production of IL-10 is an important self-regulatory function of CD4(+) T lymphocytes.
Collapse
|
33
|
Jailwala P, Waukau J, Glisic S, Jana S, Ehlenbach S, Hessner M, Alemzadeh R, Matsuyama S, Laud P, Wang X, Ghosh S. Apoptosis of CD4+ CD25(high) T cells in type 1 diabetes may be partially mediated by IL-2 deprivation. PLoS One 2009; 4:e6527. [PMID: 19654878 PMCID: PMC2716541 DOI: 10.1371/journal.pone.0006527] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 07/02/2009] [Indexed: 01/26/2023] Open
Abstract
Background Type 1 diabetes (T1D) is a T-cell mediated autoimmune disease targeting the insulin-producing pancreatic β cells. Naturally occurring FOXP3+CD4+CD25high regulatory T cells (Tregs) play an important role in dominant tolerance, suppressing autoreactive CD4+ effector T cell activity. Previously, in both recent-onset T1D patients and β cell antibody-positive at-risk individuals, we observed increased apoptosis and decreased function of polyclonal Tregs in the periphery. Our objective here was to elucidate the genes and signaling pathways triggering apoptosis in Tregs from T1D subjects. Principal Findings Gene expression profiles of unstimulated Tregs from recent-onset T1D (n = 12) and healthy control subjects (n = 15) were generated. Statistical analysis was performed using a Bayesian approach that is highly efficient in determining differentially expressed genes with low number of replicate samples in each of the two phenotypic groups. Microarray analysis showed that several cytokine/chemokine receptor genes, HLA genes, GIMAP family genes and cell adhesion genes were downregulated in Tregs from T1D subjects, relative to control subjects. Several downstream target genes of the AKT and p53 pathways were also upregulated in T1D subjects, relative to controls. Further, expression signatures and increased apoptosis in Tregs from T1D subjects partially mirrored the response of healthy Tregs under conditions of IL-2 deprivation. CD4+ effector T-cells from T1D subjects showed a marked reduction in IL-2 secretion. This could indicate that prior to and during the onset of disease, Tregs in T1D may be caught up in a relatively deficient cytokine milieu. Conclusions In summary, expression signatures in Tregs from T1D subjects reflect a cellular response that leads to increased sensitivity to apoptosis, partially due to cytokine deprivation. Further characterization of these signaling cascades should enable the detection of genes that can be targeted for restoring Treg function in subjects predisposed to T1D.
Collapse
Affiliation(s)
- Parthav Jailwala
- The Max McGee National Research Center for Juvenile Diabetes and The Human and Molecular Genetics Center, Department of Pediatrics at the Medical College of Wisconsin and the Children's Research Institute of the Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Jill Waukau
- The Max McGee National Research Center for Juvenile Diabetes and The Human and Molecular Genetics Center, Department of Pediatrics at the Medical College of Wisconsin and the Children's Research Institute of the Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Sanja Glisic
- The Max McGee National Research Center for Juvenile Diabetes and The Human and Molecular Genetics Center, Department of Pediatrics at the Medical College of Wisconsin and the Children's Research Institute of the Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Srikanta Jana
- The Max McGee National Research Center for Juvenile Diabetes and The Human and Molecular Genetics Center, Department of Pediatrics at the Medical College of Wisconsin and the Children's Research Institute of the Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Sarah Ehlenbach
- The Max McGee National Research Center for Juvenile Diabetes and The Human and Molecular Genetics Center, Department of Pediatrics at the Medical College of Wisconsin and the Children's Research Institute of the Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Martin Hessner
- The Max McGee National Research Center for Juvenile Diabetes and The Human and Molecular Genetics Center, Department of Pediatrics at the Medical College of Wisconsin and the Children's Research Institute of the Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ramin Alemzadeh
- Children's Hospital of Wisconsin Diabetes Center, Pediatric Endocrinology and Metabolism, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Shigemi Matsuyama
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Purushottam Laud
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Xujing Wang
- Department of Physics & the Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Soumitra Ghosh
- The Max McGee National Research Center for Juvenile Diabetes and The Human and Molecular Genetics Center, Department of Pediatrics at the Medical College of Wisconsin and the Children's Research Institute of the Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
34
|
Gabrysová L, Nicolson KS, Streeter HB, Verhagen J, Sabatos-Peyton CA, Morgan DJ, Wraith DC. Negative feedback control of the autoimmune response through antigen-induced differentiation of IL-10-secreting Th1 cells. ACTA ACUST UNITED AC 2009; 206:1755-67. [PMID: 19635862 PMCID: PMC2722173 DOI: 10.1084/jem.20082118] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Regulation of the immune response to self- and foreign antigens is vitally important for limiting immune pathology associated with both infections and hypersensitivity conditions. Control of autoimmune conditions can be reinforced by tolerance induction with peptide epitopes, but the mechanism is not currently understood. Repetitive intranasal administration of soluble peptide induces peripheral tolerance in myelin basic protein (MBP)–specific TCR transgenic mice. This is characterized by the presence of anergic, interleukin (IL)-10–secreting CD4+ T cells with regulatory function (IL-10 T reg cells). The differentiation pathway of peptide-induced IL-10 T reg cells was investigated. CD4+ T cells became anergic after their second encounter with a high-affinity MBP peptide analogue. Loss of proliferative capacity correlated with a switch from the Th1-associated cytokines IL-2 and interferon (IFN)-γ to the regulatory cytokine IL-10. Nevertheless, IL-10 T reg cells retained the capacity to produce IFN-γ and concomitantly expressed T-bet, demonstrating their Th1 origin. IL-10 T reg cells suppressed dendritic cell maturation, prevented Th1 cell differentiation, and thereby created a negative feedback loop for Th1-driven immune pathology. These findings demonstrate that Th1 responses can be self-limiting in the context of peripheral tolerance to a self-antigen.
Collapse
Affiliation(s)
- Leona Gabrysová
- Department of Cellular and Molecular Medicine, University of Bristol School of Medical Sciences, Bristol BS8 1TD, England, UK.
| | | | | | | | | | | | | |
Collapse
|
35
|
Gorczynski RM, Chen Z, He W, Khatri I, Sun Y, Yu K, Boudakov I. Expression of a CD200 transgene is necessary for induction but not maintenance of tolerance to cardiac and skin allografts. THE JOURNAL OF IMMUNOLOGY 2009; 183:1560-8. [PMID: 19592654 DOI: 10.4049/jimmunol.0900200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD200, a type 2 transmembrane molecule of the Ig supergene family, can induce immunosuppression in a number of biological systems, as well as promote increased graft acceptance, following binding to its receptors (CD200Rs). Skin and cardiac allograft acceptance are readily induced in transgenic mice overexpressing CD200 under control of a doxycycline-inducible promoter, both of which are associated with increased intragraft expression of mRNAs for a number of genes associated with altered T cell subset differentiation, including GATA-3, type 2 cytokines (IL-4, IL-13), GITR, and Foxp3. Interestingly, some 12-15 days after grafting, induction of transgenic CD200 expression can be stopped (by doxycycline withdrawal), without obvious significant effect on graft survival. However, neutralization of all CD200 expression (including endogenous CD200 expression) by anti-CD200 mAb caused graft loss, as did introduction of an acute inflammatory stimulus (LPS, 10 microg/mouse, delivered by i.p. injection). We conclude that even with apparently stably accepted tissue allografts, disruption of the immunoregulatory balance by an intense inflammatory stimulus can cause graft loss.
Collapse
|
36
|
Abstract
T cells must integrate multiple environmental cues when deciding whether to mount an immunogenic or tolerogenic response. Since not all self-reactive T cells are eliminated during thymic development, mechanisms of peripheral tolerance such as T cell anergy contribute to preventing autoimmunity. Recent studies have implicated extracellular adenosine and the adenosine A(2A) receptor as playing an important role in inhibiting T cell effector function. Herein, we review the current literature regarding T cell anergy and the emerging literature implicating the A(2A) receptor as critical regulator of immune activation. Finally, we present evidence to suggest a possible role for adenosine A(2A) receptor signaling in T cell anergy.
Collapse
Affiliation(s)
- Paul E Zarek
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | | |
Collapse
|
37
|
Abstract
Recent advances in immunology have led to exciting new possibilities in medicine. We now understand how to design vaccine capable of resetting the balance between immune effector and regulatory cells in people suffering from autoimmune and allergic diseases. These approaches are effective while avoiding the side effects associated with non-specific immune suppression.
Collapse
Affiliation(s)
- David C Wraith
- Department of Cellular and Molecular Medicine, University of Bristol, Bristol, UK.
| |
Collapse
|
38
|
Satoguina JS, Adjobimey T, Arndts K, Hoch J, Oldenburg J, Layland LE, Hoerauf A. Tr1 and naturally occurring regulatory T cells induce IgG4 in B cells through GITR/GITR-L interaction, IL-10 and TGF-β. Eur J Immunol 2008; 38:3101-13. [DOI: 10.1002/eji.200838193] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
39
|
Zhu B, Symonds ALJ, Martin JE, Kioussis D, Wraith DC, Li S, Wang P. Early growth response gene 2 (Egr-2) controls the self-tolerance of T cells and prevents the development of lupuslike autoimmune disease. ACTA ACUST UNITED AC 2008; 205:2295-307. [PMID: 18779345 PMCID: PMC2556781 DOI: 10.1084/jem.20080187] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Maintaining tolerance of T cells to self-antigens is essential to avoid autoimmune disease. How self-reactive T cells are kept functionally inactive is, however, unknown. In this study, we show that early growth response gene 2 (Egr-2), a zinc-finger transcription factor, is expressed in CD44high T cells and controls their proliferation and activation. In the absence of Egr-2, CD44high, but not CD44low T cells, are hyperreactive and hyperproliferative in vivo. The accumulation of activated CD4+CD44high T cells leads to the development of a late onset lupuslike autoimmune disease characterized by the accumulation of interferon (IFN)-γ and interleukin (IL)-17–producing CD4+ T cells, loss of tolerance to nuclear antigens, massive infiltration of T cells into multiple organs and glomerulonephritis. We found that the expression of cyclin-dependent kinase inhibitor p21cip1 was impaired in Egr-2–deficient T cells, whereas the expression of IFN-γ and IL-17 in response to T cell receptor ligation was significantly increased, suggesting that Egr-2 activates the expression of genes involved in the negative regulation of T cell proliferation and inflammation. These results demonstrate that Egr-2 is an intrinsic regulator of effector T cells and controls the expansion of self-reactive T cells and development of autoimmune disease.
Collapse
Affiliation(s)
- Bo Zhu
- Institute of Cell and Molecular Science, Barts and London School of Medicine and Dentistry, University of London, London E1 2AT, England, UK
| | | | | | | | | | | | | |
Collapse
|
40
|
Collins S, Lutz MA, Zarek PE, Anders RA, Kersh GJ, Powell JD. Opposing regulation of T cell function by Egr-1/NAB2 and Egr-2/Egr-3. Eur J Immunol 2008; 38:528-36. [PMID: 18203138 DOI: 10.1002/eji.200737157] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
TCR-induced NF-AT activation leads to the up-regulation of multiple genes involved in T cell anergy. Since NF-AT is also involved in T cell activation, we have endeavored to dissect TCR-induced activating and inhibitory genetic programs. This approach revealed roles for the early growth response (Egr) family of transcription factors and the Egr coactivator/corepressor NGFI-A-binding protein (NAB)2 in regulating T cell function. TCR-induced Egr-1 and NAB2 enhance T cell function, while Egr-2 and Egr-3 inhibit T cell function. In this report, we demonstrate that Egr-2 and Egr-3 are induced by NF-AT in the absence of AP-1, while Egr-1 and NAB2 both require AP-1-mediated transcription. Our data suggest that Egr-3 is upstream of Egr-2, and that mechanistically Egr-2 and Egr-3 suppress Egr-1 and NAB2 expression. Functionally, T cells from Egr-2 and Egr-3 null mice are hyperresponsive while T cells from Egr-3 transgenic, overexpressing mice are hyporesponsive. Furthermore, an in vivo model of autoimmune pneumonitis reveals that T cells from Egr-3 null mice hasten death while Egr-3-overexpressing T cells cause less disease. Overall, our data suggest that just as the Egr/NAB network of genes control cell fate in other systems, TCR-induced Egr-1, 2, 3 and NAB2 control the fate of antigen recognition in T cells.
Collapse
Affiliation(s)
- Sam Collins
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | |
Collapse
|
41
|
Marzo N, Ortega S, Stratmann T, García A, Ríos M, Giménez A, Gomis R, Mora C. Cyclin-dependent kinase 4 hyperactivity promotes autoreactivity in the immune system but protects pancreatic cell mass from autoimmune destruction in the nonobese diabetic mouse model. THE JOURNAL OF IMMUNOLOGY 2008; 180:1189-98. [PMID: 18178859 DOI: 10.4049/jimmunol.180.2.1189] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cyclin-dependent kinase 4 (Cdk4) plays a central role in perinatal pancreatic beta cell replication, thus becoming a potential target for therapeutics in autoimmune diabetes. Its hyperactive form, Cdk4R24C, causes beta cell hyperplasia without promoting hypoglycemia in a nonautoimmune-prone mouse strain. In this study, we explore whether beta cell hyperproliferation induced by the Cdk4R24C mutation balances the autoimmune attack against beta cells inherent to the NOD genetic background. To this end, we backcrossed the Cdk4R24C knockin mice, which have the Cdk4 gene replaced by the Cdk4R24C mutated form, onto the NOD genetic background. In this study, we show that NOD/Cdk4R24C knockin mice exhibit exacerbated diabetes and insulitis, and that this exacerbated diabetic phenotype is solely due to the hyperactivity of the NOD/Cdk4R24C immune repertoire. Thus, NOD/Cdk4R24C splenocytes confer exacerbated diabetes when adoptively transferred into NOD/SCID recipients, compared with NOD/wild-type (WT) donor splenocytes. Accordingly, NOD/Cdk4R24C splenocytes show increased basal proliferation and higher activation markers expression compared with NOD/WT splenocytes. However, to eliminate the effect of the Cdk4R24C mutation specifically in the lymphocyte compartment, we introduced this mutation into NOD/SCID mice. NOD/SCID/Cdk4R24C knockin mice develop beta cell hyperplasia spontaneously. Furthermore, NOD/SCID/Cdk4R24C knockin females that have been adoptively transferred with NOD/WT splenocytes are more resistant to autoimmunity than NOD/SCID WT female. Thus, the Cdk4R24C mutation opens two avenues in the NOD model: when expressed specifically in beta cells, it provides a new potential strategy for beta cell regeneration in autoimmune diabetes, but its expression in the immune repertoire exacerbates autoimmunity.
Collapse
Affiliation(s)
- Nuria Marzo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer and University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Sumpter TL, Payne KK, Wilkes DS. Regulation of the NFAT pathway discriminates CD4+CD25+ regulatory T cells from CD4+CD25- helper T cells. J Leukoc Biol 2007; 83:708-17. [PMID: 18032693 DOI: 10.1189/jlb.0507321] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
CD4(+)CD25(+) regulatory T cells (Tregs) are potent modulators of immune responses. The transcriptional program distinguishing Tregs from the CD4(+)CD25(-) Th cells is unclear. NFAT, a key transcription factor, is reported to interact with forkhead box p3, allowing inhibitory and activating signals in T cells. In the current study, we hypothesize that distinctive NFAT regulation in Tregs as compared with Th cells, may contribute to specific functions of these cells. Tregs express basal levels of cytoplasmic NFATc1 and NFATc2. In contrast to Th cells, anti-CD3-mediated T cell activation did not induce nuclear translocation of NFATc1 or NFATc2 in Tregs. This effect was associated with altered regulation for NFAT in Tregs that included reduced calcium flux, diminished calcineurin activation, and increased activity of glycogen synthase kinase-3beta, a negative regulatory kinase for NFAT in Tregs relative to Th cells. These data suggested that NFAT inhibition in Th cells may induce regulatory function. Indeed, pharmacologically mediated NFAT inhibition induced Th cells to function as Tregs, an effect that was mediated by induction of membrane-bound TGF-beta on Th cells. Collectively, these data suggest that maintaining NFAT at basal levels is a part of the transcriptional program required for Tregs.
Collapse
Affiliation(s)
- Tina L Sumpter
- Department of Microbiology and Immunology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA
| | | | | |
Collapse
|
43
|
Pallandre JR, Brillard E, Créhange G, Radlovic A, Remy-Martin JP, Saas P, Rohrlich PS, Pivot X, Ling X, Tiberghien P, Borg C. Role of STAT3 in CD4+CD25+FOXP3+Regulatory Lymphocyte Generation: Implications in Graft-versus-Host Disease and Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 2007; 179:7593-604. [DOI: 10.4049/jimmunol.179.11.7593] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
44
|
Cabbage SE, Huseby ES, Sather BD, Brabb T, Liggitt D, Goverman J. Regulatory T cells maintain long-term tolerance to myelin basic protein by inducing a novel, dynamic state of T cell tolerance. THE JOURNAL OF IMMUNOLOGY 2007; 178:887-96. [PMID: 17202350 DOI: 10.4049/jimmunol.178.2.887] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The pathogenesis of multiple sclerosis involves a breakdown in T cell tolerance to myelin proteins like myelin basic protein (MBP). Most MBP-specific T cells are eliminated by central tolerance in adult mice, however, the developmentally regulated expression of MBP allows MBP-specific thymocytes in young mice to escape negative selection. It is not known how these T cells that encounter MBP for the first time in the periphery are regulated. We show that naive MBP-specific T cells transferred into T cell-deficient mice induce severe autoimmunity. Regulatory T cells prevent disease, however, suppression of the newly transferred MBP-specific T cells is abrogated by activating APCs in vivo. Without APC activation, MBP-specific T cells persist in the periphery of protected mice but do not become anergic, raising the question of how long-term tolerance can be maintained if APCs presenting endogenous MBP become activated. Our results demonstrate that regulatory T cells induce naive MBP-specific T cells responding to nonactivated APCs to differentiate into a unique, tolerized state with the ability to produce IL-10 and TGF-beta1 in response to activated, but not nonactivated, APCs presenting MBP. This tolerant response depends on continuous activity of regulatory T cells because, in their absence, these uniquely tolerized MBP-specific T cells can again induce autoimmunity.
Collapse
Affiliation(s)
- Sarah E Cabbage
- Molecular and Cellular Biology Program, Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | |
Collapse
|
45
|
Serfling E, Klein-Hessling S, Palmetshofer A, Bopp T, Stassen M, Schmitt E. NFAT transcription factors in control of peripheral T cell tolerance. Eur J Immunol 2007; 36:2837-43. [PMID: 17039563 DOI: 10.1002/eji.200536618] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The Ca++-regulated calcineurin/NFAT cascade is one of the crucial signalling pathways that controls adaptive immunity. However, a number of novel experimental data suggest that, in addition to their role in T cell activation, NFATc transcription factors play also a decisive role in the generation of peripheral tolerance against self-antigens. This function of NFATc factors is mediated by controlling activation-induced cell death and clonal anergy of T helper cells and the activity of regulatory T cells. The multi-functional role of NFATc proteins characterize these transcription factors as key regulators of immunological tolerance and, if dysregulated, of development of autoimmune diseases.
Collapse
Affiliation(s)
- Edgar Serfling
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany.
| | | | | | | | | | | |
Collapse
|
46
|
Peng ZC, Cai X, Zhang YG, Kong DS, Guo HS, Liang W, Tang QQ, Song HY, Ma D. A novel anti-tissue factor monoclonal antibody with anticoagulant potency derived from synthesized multiple antigenic peptide through blocking FX combination with TF. Thromb Res 2007; 121:85-93. [PMID: 17573098 DOI: 10.1016/j.thromres.2007.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 02/11/2007] [Accepted: 03/02/2007] [Indexed: 10/23/2022]
Abstract
Tissue factor (TF) has been implicated in the pathogenesis of various thrombotic disorders. Monoclonal antibodies (mAb) that specifically target TF may have potential as antithrombotic therapy. We designed a unique TF peptide (TFP) that was specific for the binding site to factor X (FX). This peptide was used to develop TF mAb that block the coagulation cascade by interfering with the combination of FX with the TF/FVIIa complex. Chemically synthesized TFP coupled to polylysine matrix was used as multiple antigenic peptide (TF-MAP) and this was used to immunize Balb/c mice for the preparation of hybridomas. One hybridoma cell line released an antibody, named TF4A12, which had high anticoagulant potency (by dilute prothrombin time assay). Western blotting showed that TF4A12 could bind TF-MAP and the soluble TF extracellular domain (sTF(1-219)). Results of FX activation assay and amidolytic activity assay showed that the anticoagulant ability of TF4A12 is due to blocking FX, but not FVII, binding to TF. Our study identified an efficient method of developing TF mAb that could block the coagulation cascade.
Collapse
Affiliation(s)
- Zhuo-Chun Peng
- Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Fudan University, 200032, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Roord ST, Zonneveld-Huijssoon E, Le T, Yung GP, Koffeman E, Ronaghy A, Ghahramani N, Lanza P, Billetta R, Prakken BJ, Albani S. Modulation of T cell function by combination of epitope specific and low dose anticytokine therapy controls autoimmune arthritis. PLoS One 2006; 1:e87. [PMID: 17183718 PMCID: PMC1762388 DOI: 10.1371/journal.pone.0000087] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Accepted: 11/09/2006] [Indexed: 12/16/2022] Open
Abstract
Innate and adaptive immunity contribute to the pathogenesis of autoimmune arthritis by generating and maintaining inflammation, which leads to tissue damage. Current biological therapies target innate immunity, eminently by interfering with single pro-inflammatory cytokine pathways. This approach has shown excellent efficacy in a good proportion of patients with Rheumatoid Arthritis (RA), but is limited by cost and side effects. Adaptive immunity, particularly T cells with a regulatory function, plays a fundamental role in controlling inflammation in physiologic conditions. A growing body of evidence suggests that modulation of T cell function is impaired in autoimmunity. Restoration of such function could be of significant therapeutic value. We have recently demonstrated that epitope-specific therapy can restore modulation of T cell function in RA patients. Here, we tested the hypothesis that a combination of anti-cytokine and epitope-specific immunotherapy may facilitate the control of autoimmune inflammation by generating active T cell regulation. This novel combination of mucosal tolerization to a pathogenic T cell epitope and single low dose anti-TNFα was as therapeutically effective as full dose anti-TNFα treatment. Analysis of the underlying immunological mechanisms showed induction of T cell immune deviation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/therapy
- Autoimmunity
- Base Sequence
- Chaperonin 60/genetics
- Chaperonin 60/immunology
- Cytokines/antagonists & inhibitors
- Cytokines/genetics
- Cytokines/metabolism
- DNA Primers/genetics
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Etanercept
- Hindlimb/pathology
- Humans
- Immunity, Mucosal
- Immunoglobulin G/administration & dosage
- Immunotherapy/methods
- Immunotherapy, Adoptive
- Male
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Rats
- Rats, Inbred Lew
- Receptors, Tumor Necrosis Factor/administration & dosage
- T-Lymphocytes/immunology
- T-Lymphocytes, Regulatory/immunology
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Sarah T.A. Roord
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatric Immunology, University Medical Center Utrecht, Wilhelmina Children's HospitalUtrecht, The Netherlands
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Evelien Zonneveld-Huijssoon
- Department of Pediatric Immunology, University Medical Center Utrecht, Wilhelmina Children's HospitalUtrecht, The Netherlands
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Tho Le
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Gisella Puga Yung
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Eva Koffeman
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Arash Ronaghy
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
| | - Negar Ghahramani
- Androclus TherapeuticsSan Diego, California, United States of America
| | - Paola Lanza
- Androclus TherapeuticsSan Diego, California, United States of America
| | - Rosario Billetta
- Androclus TherapeuticsSan Diego, California, United States of America
| | - Berent J. Prakken
- Department of Pediatric Immunology, University Medical Center Utrecht, Wilhelmina Children's HospitalUtrecht, The Netherlands
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Salvatore Albani
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Androclus TherapeuticsSan Diego, California, United States of America
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
48
|
Cobbold SP. The hidden truth about gene expression in Tregs: Is it what you don't see that counts? Eur J Immunol 2006; 36:1360-3. [PMID: 16708406 DOI: 10.1002/eji.200636171] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A population of IL-10 secreting regulatory T cells (PI-Tregs) can be induced in a mouse TCR transgenic model of experimental autoimmune encephalomyelitis (EAE) by intranasal administration of the N-terminal peptide of myelin basic protein. A paper in this issue of the European Journal of Immunology identifies patterns of gene expression that distinguish PI-Treg from naïve T cells after activation in vivo. PI-Tregs are anergic and the expression of egr2 may be involved in regulation of the cell cycle. The surprising expression of the Th1 determining gene T-bet in PI-Tregs may be functionally relevant as transfection of T-bet into a hybridoma induced some genes associated with regulatory T cells, such as tlr2 and gzmb. In this commentary, PI-Tregs are considered in comparison to other Tregs, such as the natural CD4(+)CD25(+) and Tr1 cells, and there seems to be considerable overlap in gene expression between all three Treg populations, but also with both Th1 and Th2 cells. It is suggested that regulatory activity may be a common feature of all activated T cells and that a defining principle of a Treg is the lack of effector functions due to a partial or incomplete differentiation to either Th1 or Th2.
Collapse
|