1
|
Laukova M, Glatman Zaretsky A. Regulatory T cells as a therapeutic approach for inflammatory bowel disease. Eur J Immunol 2023; 53:e2250007. [PMID: 36562391 PMCID: PMC10107179 DOI: 10.1002/eji.202250007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/20/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Foxp3+ T regulatory (Treg) cells suppress inflammation and are essential for maintaining tissue homeostasis. A growing appreciation of tissue-specific Treg functions has built interest in leveraging the endogenous suppressive mechanisms of these cells into cellular therapeutics in organ-specific diseases. Notably, Treg cells play a critical role in maintaining the intestinal environment. As a barrier site, the gut requires Treg cells to mediate interactions with the microbiota, support barrier integrity, and regulate the immune system. Without fully functional Treg cells, intestinal inflammation and microbial dysbiosis ensue. Thus, there is a particular interest in developing Treg cellular therapies for intestinal inflammatory disease, such as inflammatory bowel disease (IBD). This article reviews some of the critical pathways that are dysregulated in IBD, Treg cell mechanisms of suppression, and the efforts and approaches in the field to develop these cells as a cellular therapy for IBD.
Collapse
|
2
|
Dong Y, Yang C, Pan F. Post-Translational Regulations of Foxp3 in Treg Cells and Their Therapeutic Applications. Front Immunol 2021; 12:626172. [PMID: 33912156 PMCID: PMC8071870 DOI: 10.3389/fimmu.2021.626172] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Regulatory T (Treg) cells are indispensable for immune homeostasis due to their roles in peripheral tolerance. As the master transcription factor of Treg cells, Forkhead box P3 (Foxp3) strongly regulates Treg function and plasticity. Because of this, considerable research efforts have been directed at elucidating the mechanisms controlling Foxp3 and its co-regulators. Such work is not only advancing our understanding on Treg cell biology, but also uncovering novel targets for clinical manipulation in autoimmune diseases, organ transplantation, and tumor therapies. Recently, many studies have explored the post-translational regulation of Foxp3, which have shown that acetylation, phosphorylation, glycosylation, methylation, and ubiquitination are important for determining Foxp3 function and plasticity. Additionally, some of these targets have been implicated to have great therapeutic values. In this review, we will discuss emerging evidence of post-translational regulations on Foxp3 in Treg cells and their exciting therapeutic applications.
Collapse
Affiliation(s)
- Yi Dong
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Cuiping Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fan Pan
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| |
Collapse
|
3
|
Dong Y, Pan F. Ubiquitin-Dependent Regulation of Treg Function and Plasticity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:63-80. [PMID: 33523443 DOI: 10.1007/978-981-15-6407-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
As an indispensable part of peripheral tolerance, regulatory T (Treg) cells play an important role in immune homeostasis by suppressing other immune cells. Behind this function is a complex network of transcription factors and signaling cascades that regulates the function and plasticity of regulatory T cells. Among these, Forkhead box P3 (Foxp3) is considered as the master transcription factor, and its stability will influence the function and viability of Treg cells. Because of this, understanding the mechanisms that regulate Foxp3 and its co-regulators will provide more understanding to Treg cells and uncover more targets to manipulate Treg cells in treating autoimmune diseases, organ transplantation, and tumor. Interestingly, several recent studies show that ubiquitin-dependent pathways are important regulators of Foxp3, which suggest both great scientific and therapeutic values. In this chapter, we cover emerging evidence of ubiquitin-dependent, posttranslational regulation of Treg function and plasticity.
Collapse
Affiliation(s)
- Yi Dong
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fan Pan
- Center for Cancer Immunology Research, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| |
Collapse
|
4
|
Targeting Neuropilin-1 Suppresses the Stability of CD4 + CD25 + Regulatory T Cells via the NF-κB Signaling Pathway in Sepsis. Infect Immun 2021; 89:IAI.00399-20. [PMID: 33139385 DOI: 10.1128/iai.00399-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/26/2020] [Indexed: 01/19/2023] Open
Abstract
Neuropilin-1 (Nrp-1) contributes to maintaining the stability of CD4+ CD25+ regulatory T cells (Tregs). We investigated the impact of Nrp-1 on the stability of CD4+ CD25+ Tregs, and the underlying signaling pathways, in a model of sepsis. Splenic CD4+ CD25+ Tregs were either treated with anti-Nrp-1, transfected to silence Nrp-1 and inhibitor of NF-κB kinase subunit beta (IKKβ), or administered ammonium pyrrolidine dithiocarbamate (PDTC), followed by recombinant semaphorin 3A (rSema3A), in a simulation of sepsis. After the creation of a sepsis model in mice, anti-Nrp-1 was administered. The expression of the gene encoding forkhead box protein P-3 foxp3-Treg-specific demethylated region (foxp3-TSDR), the apoptosis rate, the expression of Foxp-3, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), and transforming growth factor β1 (TGF-β1), interleukin 10 (IL-10) and TGF-β1 secretion, and the NF-κB signaling activity of CD4+ CD25+ Tregs were determined. Sepsis simulation with or without rSema3A increased the stability of CD4+ CD25+ Tregs, including an increase in the expression of Foxp-3, CTLA-4, and TGF-β1, decreases in apoptosis and the methylation of foxp3-TSDR, increases in the secretion of TGF-β1 and IL-10, and an increase in the immunosuppressive effect on CD4+ T lymphocytes. Silencing of Nrp-1 or anti-Nrp-1 treatment abrogated lipopolysaccharide (LPS) stimulation with or without an rSema3A-mediated effect. Sepsis simulation increased the DNA-binding activity of NF-κB, as well as the ratios of phosphorylated IKKβ (p-IKKβ) to IKKβ and p-P65 to P65 in vitro and vivo Silencing of IKKβ expression or PDTC treatment suppressed the stability of CD4+ CD25+ Tregs in LPS-induced sepsis. Weakening Nrp-1 reduced the stability of CD4+ CD25+ Tregs by regulating the NF-κB signaling pathway; thus, Nrp-1 could be a new target for immunoregulation in sepsis.
Collapse
|
5
|
Moyé S, Bormann T, Maus R, Sparwasser T, Sandrock I, Prinz I, Warnecke G, Welte T, Gauldie J, Kolb M, Maus UA. Regulatory T Cells Limit Pneumococcus-Induced Exacerbation of Lung Fibrosis in Mice. THE JOURNAL OF IMMUNOLOGY 2020; 204:2429-2438. [PMID: 32213566 DOI: 10.4049/jimmunol.1900980] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 03/02/2020] [Indexed: 12/28/2022]
Abstract
Patients with idiopathic pulmonary fibrosis (IPF) can experience life-threatening episodes of acute worsening of their disease, termed acute exacerbation of IPF, which may be caused by bacterial and/or viral infections. The potential for regulatory T cells (Tregs) to limit disease progression in bacterially triggered fibrosis exacerbation has not been explored so far. In the current study, we show that the number of Tregs was significantly increased in mice with established AdTGF-β1-induced lung fibrosis and further increased in mice with pneumococcal infection-induced lung fibrosis exacerbation. Diphtheria toxin-induced depletion of Tregs significantly worsened infection-induced fibrosis exacerbation as determined by increased lung collagen deposition, lung histology, and elevated pulmonary Th1/Th2 cytokine levels. Conversely, IL-2 complex-induced Treg expansion in wild-type mice with established lung fibrosis completely inhibited pneumococcal infection-induced fibrosis exacerbation as efficaciously as antibiotic treatment while preserving lung antibacterial immunity in mice. Collectively, these findings demonstrate the efficacy of Tregs as "silencers," suppressing infection-induced exacerbation of lung fibrosis in mice, and their expansion may offer a novel adjunctive treatment to limit acute exacerbations in patients with IPF.
Collapse
Affiliation(s)
- Steffi Moyé
- Division of Experimental Pneumology, Hannover Medical School, Hannover 30625, Germany
| | - Tina Bormann
- Division of Experimental Pneumology, Hannover Medical School, Hannover 30625, Germany
| | - Regina Maus
- Division of Experimental Pneumology, Hannover Medical School, Hannover 30625, Germany
| | - Tim Sparwasser
- Department of Medical Microbiology and Hygiene, University Medical Center Mainz, Mainz 55131, Germany
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover 30625, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover 30625, Germany
| | - Gregor Warnecke
- Division of Cardiac, Thoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover 30625, Germany.,Partner Site BREATH, German Center for Lung Research, Hannover 30625, Germany
| | - Tobias Welte
- Partner Site BREATH, German Center for Lung Research, Hannover 30625, Germany.,Clinic for Pneumology, Hannover Medical School, Hannover 30625, Germany; and
| | | | | | - Ulrich A Maus
- Division of Experimental Pneumology, Hannover Medical School, Hannover 30625, Germany; .,Partner Site BREATH, German Center for Lung Research, Hannover 30625, Germany
| |
Collapse
|
6
|
Macciò A, Madeddu C. Blocking inflammation to improve immunotherapy of advanced cancer. Immunology 2019; 159:357-364. [PMID: 31821533 DOI: 10.1111/imm.13164] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/19/2022] Open
Abstract
The ability to induce functional reprogramming of regulatory T (Treg) cells in the tumor microenvironment is an extremely important therapeutic opportunity. However, when discussing such an approach, the opposing effect that the activation of the Treg cell compartments may have in inducing the immune inflammatory response and its link with the efficacy of immunotherapy should be considered. In fact, Treg reprogramming has a dual effect: immediate, with mechanisms that activate immunosurveillance, and late, mediated by the macrophage activation that yields an inflammatory status that is deleterious for the antineoplastic efficiency of the immune system response. Persistence of the inflammatory response is associated with specific changes of oxidative and glycolytic metabolic pathways that interfere with conventional T-cell activation and function and may be one of the reasons for the failure of immunotherapy in advanced cancer patients. Therefore, in addition to modulating Treg cell action, the combined use of drugs able to block chronic inflammation mediated mainly by macrophages, to counteract the oxidative stress, and to positively regulate the metabolic derangements, could improve the effectiveness of modern immunotherapy. In conclusion, reprogramming of Treg cells may be an appropriate strategy for treating early stages of neoplastic diseases, whereas other immunosuppressive mechanisms should be the target of a combined immunotherapy approach in more advanced phases of cancer.
Collapse
Affiliation(s)
- Antonio Macciò
- Department of Gynecologic Oncology, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
7
|
Abstract
Regulatory T (Treg) cells are a distinct subset of CD4+ T cells. Instead of triggering adaptive immunity, they suppress immune responses. Small numbers of Treg cells reside within lymphoid organs and peripheral tissues, but their contribution to immune tolerance is so significant that defects in Treg cell function cause catastrophic immune disorders. Since they were first discovered 20 years ago, efforts have been made to understand the differences in developmental processes between Treg cells and conventional T cells that determine the ultimate fate of the overall T-cell population. Transcription factor Foxp3 is crucial for Treg cell differentiation, but it is not the whole story. Owing to recent advances in Treg cell research, we are now on the verge of appreciating the comprehensive mechanisms underlying Treg cell generation. Here, we discuss major discoveries, active study topics and remaining questions regarding Treg cell development.
Collapse
Affiliation(s)
- Wonyong Lee
- Department of Life Science, Sogang University, Mapo-gu, Seoul 04107, Korea
| | - Gap Ryol Lee
- Department of Life Science, Sogang University, Mapo-gu, Seoul 04107, Korea
| |
Collapse
|
8
|
Phillips BE, Garciafigueroa Y, Trucco M, Giannoukakis N. Clinical Tolerogenic Dendritic Cells: Exploring Therapeutic Impact on Human Autoimmune Disease. Front Immunol 2017; 8:1279. [PMID: 29075262 PMCID: PMC5643419 DOI: 10.3389/fimmu.2017.01279] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022] Open
Abstract
Tolerogenic dendritic cell (tDC)-based clinical trials for the treatment of autoimmune diseases are now a reality. Clinical trials are currently exploring the effectiveness of tDC to treat autoimmune diseases of type 1 diabetes mellitus, rheumatoid arthritis, multiple sclerosis (MS), and Crohn's disease. This review will address tDC employed in current clinical trials, focusing on cell characteristics, mechanisms of action, and clinical findings. To date, the publicly reported human trials using tDC indicate that regulatory lymphocytes (largely Foxp3+ T-regulatory cell and, in one trial, B-regulatory cells) are, for the most part, increased in frequency in the circulation. Other than this observation, there are significant differences in the major phenotypes of the tDC. These differences may affect the outcome in efficacy of recently launched and impending phase II trials. Recent efforts to establish a catalog listing where tDC converge and diverge in phenotype and functional outcome are an important first step toward understanding core mechanisms of action and critical "musts" for tDC to be therapeutically successful. In our view, the most critical parameter to efficacy is in vivo stability of the tolerogenic activity over phenotype. As such, methods that generate tDC that can induce and stably maintain immune hyporesponsiveness to allo- or disease-specific autoantigens in the presence of powerful pro-inflammatory signals are those that will fare better in primary endpoints in phase II clinical trials (e.g., disease improvement, preservation of autoimmunity-targeted tissue, allograft survival). We propose that pre-treatment phenotypes of tDC in the absence of functional stability are of secondary value especially as such phenotypes can dramatically change following administration, especially under dynamic changes in the inflammatory state of the patient. Furthermore, understanding the outcomes of different methods of cell delivery and sites of delivery on functional outcomes, as well as quality control variability in the functional outcomes resulting from the various approaches of generating tDC for clinical use, will inform more standardized ex vivo generation methods. An understanding of these similarities and differences, with a reference point the large number of naturally occurring tDC populations with different immune profiles described in the literature, could explain some of the expected and unanticipated outcomes of emerging tDC clinical trials.
Collapse
Affiliation(s)
- Brett Eugene Phillips
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Yesica Garciafigueroa
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Massimo Trucco
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Nick Giannoukakis
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
9
|
CD4(+)CD25(hi)Foxp3(+) Cells Exacerbate Bleomycin-Induced Pulmonary Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2008-2020. [PMID: 27317904 DOI: 10.1016/j.ajpath.2016.03.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/16/2016] [Accepted: 03/23/2016] [Indexed: 01/20/2023]
Abstract
Idiopathic pulmonary fibrosis is a fatal lung disease with a median survival of 2 to 5 years. A decade of studies has downplayed inflammation contributing to its pathogenesis. However, these studies preceded the discovery of regulatory T cells (Tregs) and all of their functions. On the basis of human studies demonstrating Tregs can decrease graft-versus-host disease and vasculitides, there is consideration of their use to treat idiopathic pulmonary fibrosis. We hypothesized that Treg therapy would attenuate the fibroplasia involved in a preclinical murine model of pulmonary fibrosis. IL-2 complex was used in vivo to expand CD4(+)CD25(hi)Foxp3(+) cells in the lung during intratracheal bleomycin challenge; however, this unexpectedly led to an increase in lung fibrosis. More important, this increase in fibrosis was a lymphocyte-dependent process. We corroborated these results using a CD4(+)CD25(hi)Foxp3(+) cellular-based therapy. Mechanistically, we demonstrated that CD4(+)CD25(hi)Foxp3(+) cells undergo alterations during bleomycin challenge and the IL-2 complex had no effect on profibrotic (eg, transforming growth factor-β) or type 17 immune response cytokines; however, there was a marked down-regulation of the type 1 and augmentation of the type 2 immune response cytokines from the lungs. Collectively, our animal studies show that a specific lung injury can induce Treg alterations, which can augment pulmonary fibrosis.
Collapse
|
10
|
Ronaldson A, Gazali AM, Zalli A, Kaiser F, Thompson SJ, Henderson B, Steptoe A, Carvalho L. Increased percentages of regulatory T cells are associated with inflammatory and neuroendocrine responses to acute psychological stress and poorer health status in older men and women. Psychopharmacology (Berl) 2016; 233:1661-8. [PMID: 25678193 PMCID: PMC4828497 DOI: 10.1007/s00213-015-3876-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/21/2015] [Indexed: 01/05/2023]
Abstract
RATIONALE The percentage of regulatory T cells (TRegs)-a subtype of T lymphocyte that suppresses the immune response-appears to be reduced in a number of stress-related diseases. The role of the TReg in stress-disease pathways has not yet been investigated. OBJECTIVES The aim of the study was to investigate the association between biological responsivity to acute psychosocial stress and the percentage of TRegs in healthy older adults. The secondary purpose was to measure the associations between TReg percentage and psychological and physical well-being in the participants. METHODS Salivary cortisol and plasma interleukin (IL)-6 samples were obtained from 121 healthy older men and women from the Whitehall II cohort following acute psychophysiological stress testing. Three years later at a follow-up visit, we measured TReg percentages and psychological and physical well-being were recorded using the Short Form 36 Health Survey and the Center for Epidemiologic Studies Depression Scale. RESULTS Blunted cortisol responses (p = 0.004) and elevated IL-6 responses (p = 0.027) to acute psychophysiological stress were associated with greater TReg percentage independently of age, sex, BMI, smoking status, employment grade, time of testing, and baseline measures of cortisol and IL-6, respectively. Percentage of TRegs was associated cross-sectionally with lower physical (p = 0.043) and mental health status (p = 0.008), and higher levels of depressive symptoms (p = 0.002), independently of covariates. CONCLUSIONS Increased levels of TRegs may act as a defence against increased inflammation and may be a pre-indication for chronically stressed individuals on the cusp of clinical illness.
Collapse
Affiliation(s)
- Amy Ronaldson
- Department of Epidemiology and Public Health, University College London, 1-19, Torrington Place, London, WC1E 6BT, UK.
| | - Ahmad M. Gazali
- Department of Academic Rheumatology, Division of Immunology, Infection and Inflammatory Disease, School of Medicine, Kings College London, Guy’s Campus, London, SE1 1UL UK
| | - Argita Zalli
- Department of Epidemiology and Public Health, University College London, 1-19, Torrington Place, London, WC1E 6BT UK
| | - Frank Kaiser
- Department of Microbial Diseases, UCL-Eastman Dental Institute, London, WC1X 8LD UK
| | - Stephen J. Thompson
- Department of Academic Rheumatology, Division of Immunology, Infection and Inflammatory Disease, School of Medicine, Kings College London, Guy’s Campus, London, SE1 1UL UK
| | - Brian Henderson
- Department of Microbial Diseases, UCL-Eastman Dental Institute, London, WC1X 8LD UK
| | - Andrew Steptoe
- Department of Epidemiology and Public Health, University College London, 1-19, Torrington Place, London, WC1E 6BT UK
| | - Livia Carvalho
- Department of Epidemiology and Public Health, University College London, 1-19, Torrington Place, London, WC1E 6BT UK
| |
Collapse
|
11
|
Ye L, Goodall JC, Zhang L, Putintseva EV, Lam B, Jiang L, Liu W, Yin J, Lin L, Li T, Wu X, Yeo G, Shugay M, Chudakov DM, Gaston H, Xu H. TCR usage, gene expression and function of two distinct FOXP3
+
Treg subsets within CD4
+
CD25
hi
T cells identified by expression of CD39 and CD45RO. Immunol Cell Biol 2015; 94:293-305. [PMID: 26467610 DOI: 10.1038/icb.2015.90] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/29/2015] [Accepted: 09/29/2015] [Indexed: 02/08/2023]
Affiliation(s)
- Lingying Ye
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| | - Jane C Goodall
- Department of Medicine, School of Clinical Medicine, Addenbrookes Hospital, University of Cambridge Cambridge UK
| | - Libin Zhang
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| | - Ekaterina V Putintseva
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences Moscow Russian Federation
| | - Brian Lam
- University of Cambridge Metabolic Research Labs, Institute of Metabolic Science, Addenbrookes Hospital Cambridge UK
| | - Lei Jiang
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| | - Wei Liu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| | - Jian Yin
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| | - Li Lin
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| | - Ting Li
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| | - Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| | - Giles Yeo
- University of Cambridge Metabolic Research Labs, Institute of Metabolic Science, Addenbrookes Hospital Cambridge UK
| | - Mikhail Shugay
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences Moscow Russian Federation
- Pirogov Russian National Research Medical University Moscow Russia
| | - Dmitriy M Chudakov
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences Moscow Russian Federation
- Pirogov Russian National Research Medical University Moscow Russia
| | - Hill Gaston
- Department of Medicine, School of Clinical Medicine, Addenbrookes Hospital, University of Cambridge Cambridge UK
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Shanghai China
| |
Collapse
|
12
|
Ma S, Wang Q, Ma X, Wu L, Guo F, Ji H, Liu F, Zhao Y, Qin G. FoxP3 in papillary thyroid carcinoma induces NIS repression through activation of the TGF-β1/Smad signaling pathway. Tumour Biol 2015; 37:989-98. [PMID: 26264613 DOI: 10.1007/s13277-015-3848-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/24/2015] [Indexed: 12/13/2022] Open
Abstract
Forkhead box P3 (FoxP3) expression in papillary thyroid carcinoma (PTC) is associated with resistance to radioiodine treatment. The sodium iodine symporter (NIS) is a plasma membrane glycoprotein, the repression of which may render the tumor refractive to radioiodine therapy. In this study, samples from 90 PTCs as well as 40 normal thyroid tissues were examined for FoxP3 and NIS by immunohistochemistry and real-time PCR. We found that FoxP3 was associated with decreased NIS expression. Lentiviral-mediated FoxP3-overexpressing cells were constructed and real-time PCR and western blotting were performed to evaluate the expression of NIS. Meanwhile, key members of the transforming growth factor-β1 (TGF-β1) pathway were explored by ELISA and immunofluorescence and a neutralizing TGF-β1 antibody was used to block activity. In vitro, FoxP3 overexpression significantly reduced NIS transcript and protein levels and the TGF-β1 pathway was activated. However, treatment with neutralizing TGF-β1 antibody partially abrogated FoxP3-induced NIS repression. These findings suggest that FoxP3 could compromise NIS expression by inducing TGF-β1.
Collapse
Affiliation(s)
- Siyuan Ma
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qingzhu Wang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaojun Ma
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Lina Wu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Feng Guo
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hongfei Ji
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fei Liu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yanyan Zhao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Guijun Qin
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China.
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
13
|
Schaer DA, Budhu S, Liu C, Bryson C, Malandro N, Cohen A, Zhong H, Yang X, Houghton AN, Merghoub T, Wolchok JD. GITR pathway activation abrogates tumor immune suppression through loss of regulatory T cell lineage stability. Cancer Immunol Res 2015; 1:320-31. [PMID: 24416730 DOI: 10.1158/2326-6066.cir-13-0086] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Ligation of GITR (glucocorticoid-induced tumor necrosis factor (TNF) receptor-related gene, or TNFRSF18) by agonist antibody has recently entered into early phase clinical trials for the treatment of advanced malignancies. Although the ability of GITR modulation to induce tumor regression is well-documented in preclinical studies, the underlying mechanisms of action, particularly its effects on CD4(+)foxp3(+) regulatory T cells (Treg), have not been fully elucidated. We have previously demonstrated that GITR ligation in vivo by agonist antibody DTA-1 causes a >50% reduction of intra-tumor Treg with down modulation of Foxp3 expression. Here we show that the loss of Foxp3 is tumor-dependent. Adoptively-transferred Foxp3(+)Treg from tumor-bearing animals lose Foxp3 expression in the host when treated with DTA-1, whereas Treg from naïve mice maintain Foxp3 expression. GITR ligation also alters the expression of various transcription factors and cytokines important for Treg function. Complete Foxp3 loss in intra-tumor Treg correlates with a dramatic decrease in Helios expression and is associated with the upregulation of transcription factors T-Bet and Eomes. Changes in Helios correspond with a reduction in IL-10 and an increase in IFNγ expression in DTA-1-treated Treg. Together, these data show that GITR agonist antibody alters Treg lineage stability inducing an inflammatory effector T cell phenotype. The resultant loss of lineage stability causes Treg to lose their intra-tumor immune suppressive function, making the tumor susceptible to killing by tumor-specific effector CD8(+) T cells.
Collapse
Affiliation(s)
- David A Schaer
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Sadna Budhu
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Cailian Liu
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Campbell Bryson
- Weill Cornell Medical College, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Nicole Malandro
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY ; Weill Cornell Medical College, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Adam Cohen
- Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hong Zhong
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Xia Yang
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Alan N Houghton
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Taha Merghoub
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Jedd D Wolchok
- Swim Across America & Ludwig Collaborative Lab Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY ; Weill Cornell Medical College, Memorial Sloan-Kettering Cancer Center, New York, NY ; Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
14
|
Protein kinase CK2 enables regulatory T cells to suppress excessive TH2 responses in vivo. Nat Immunol 2015; 16:267-75. [PMID: 25599562 DOI: 10.1038/ni.3083] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/15/2014] [Indexed: 12/12/2022]
Abstract
The quality of the adaptive immune response depends on the differentiation of distinct CD4(+) helper T cell subsets, and the magnitude of an immune response is controlled by CD4(+)Foxp3(+) regulatory T cells (Treg cells). However, how a tissue- and cell type-specific suppressor program of Treg cells is mechanistically orchestrated has remained largely unexplored. Through the use of Treg cell-specific gene targeting, we found that the suppression of allergic immune responses in the lungs mediated by T helper type 2 (TH2) cells was dependent on the activity of the protein kinase CK2. Genetic ablation of the β-subunit of CK2 specifically in Treg cells resulted in the proliferation of a hitherto-unexplored ILT3(+) Treg cell subpopulation that was unable to control the maturation of IRF4(+)PD-L2(+) dendritic cells required for the development of TH2 responses in vivo.
Collapse
|
15
|
Barbi J, Pardoll D, Pan F. Treg functional stability and its responsiveness to the microenvironment. Immunol Rev 2014; 259:115-39. [PMID: 24712463 DOI: 10.1111/imr.12172] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Tregs) prevent autoimmunity and tissue damage resulting from excessive or unnecessary immune activation through their suppressive function. While their importance for proper immune control is undeniable, the stability of the Treg lineage has recently become a controversial topic. Many reports have shown dramatic loss of the signature Treg transcription factor Forkhead box protein 3 (Foxp3) and Treg function under various inflammatory conditions. Other recent studies demonstrate that most Tregs are extremely resilient in their expression of Foxp3 and the retention of suppressive function. While this debate is unlikely to be settled in the immediate future, improved understanding of the considerable heterogeneity within the Foxp3(+) Treg population and how Treg subsets respond to ranging environmental cues may be keys to reconciliation. In this review, we discuss the diverse mechanisms responsible for the observed stability or instability of Foxp3(+) Treg identity and function. These include transcriptional and epigenetic programs, transcript targeting, and posttranslational modifications that appear responsive to numerous elements of the microenvironment. These mechanisms for Treg functional modulation add to the discussion of Treg stability.
Collapse
Affiliation(s)
- Joseph Barbi
- Department of Oncology, Immunology and Hematopoiesis Division, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
16
|
d’Hennezel E, Piccirillo CA. Functional plasticity in human FOXP3+regulatory T cells. Hum Vaccin Immunother 2014; 8:1001-5. [DOI: 10.4161/hv.20203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
17
|
Abstract
Two opposing descriptions of so-called mesenchymal stem cells (MSCs) exist at this time. One sees MSCs as the postnatal, self-renewing, and multipotent stem cells for the skeleton. This cell coincides with a specific type of bone marrow perivascular cell. In skeletal physiology, this skeletal stem cell is pivotal to the growth and lifelong turnover of bone and to its native regeneration capacity. In hematopoietic physiology, its role as a key player in maintaining hematopoietic stem cells in their niche and in regulating the hematopoietic microenvironment is emerging. In the alternative description, MSCs are ubiquitous in connective tissues and are defined by in vitro characteristics and by their use in therapy, which rests on their ability to modulate the function of host tissues rather than on stem cell properties. Here, I discuss how the two views developed, conceptually and experimentally, and attempt to clarify the confusion arising from their collision.
Collapse
Affiliation(s)
- Paolo Bianco
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
18
|
Abstract
Regulatory T cells (Tregs) prevail as a specialized cell lineage that has a central role in the dominant control of immunological tolerance and maintenance of immune homeostasis. Thymus-derived Tregs (tTregs) and their peripherally induced counterparts (pTregs) are imprinted with unique Forkhead box protein 3 (Foxp3)-dependent and independent transcriptional and epigenetic characteristics that bestows on them the ability to suppress disparate immunological and non-immunological challenges. Thus, unidirectional commitment and the predominant stability of this regulatory lineage is essential for their unwavering and robust suppressor function and has clinical implications for the use of Tregs as cellular therapy for various immune pathologies. However, recent studies have revealed considerable heterogeneity or plasticity in the Treg lineage, acquisition of alternative effector or hybrid fates, and promotion rather than suppression of inflammation in extreme contexts. In addition, the absolute stability of Tregs under all circumstances has been questioned. Since these observations challenge the safety and efficacy of human Treg therapy, the issue of Treg stability versus plasticity continues to be enthusiastically debated. In this review, we assess our current understanding of the defining features of Foxp3(+) Tregs, the intrinsic and extrinsic cues that guide development and commitment to the Treg lineage, and the phenotypic and functional heterogeneity that shapes the plasticity and stability of this critical regulatory population in inflammatory contexts.
Collapse
Affiliation(s)
- Deepali V Sawant
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
19
|
Pan F, Barbi J. Ubiquitous points of control over regulatory T cells. J Mol Med (Berl) 2014; 92:555-69. [PMID: 24777637 DOI: 10.1007/s00109-014-1156-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/04/2014] [Accepted: 04/11/2014] [Indexed: 12/31/2022]
Abstract
Posttranslational modification by ubiquitin tagging is crucial for regulating the stability, activity and cellular localization of many target proteins involved in processes including DNA repair, cell cycle progression, protein quality control, and signal transduction. It has long been appreciated that ubiquitin-mediated events are important for certain signaling pathways leading to leukocyte activation and the stimulation of effector function. Now it is clear that the activities of molecules and pathways central to immune regulation are also modified and controlled by ubiquitin tagging. Among the mechanisms of immune control, regulatory T cells (or Tregs) are themselves particularly sensitive to such regulation. E3 ligases and deubiquitinases both influence Tregs through their effects on the signaling pathways pertinent to these cells or through the direct, posttranslational regulation of Foxp3. In this review, we will summarize and discuss several examples of ubiquitin-mediated control over multiple aspects of Treg biology including the generation, function and phenotypic fidelity of these cells. Fully explored and exploited, these potential opportunities for Treg modulation may lead to novel immunotherapies for both positive and negative fine-tuning of immune restraint.
Collapse
Affiliation(s)
- Fan Pan
- Immunology and Hematopoiesis Division, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA,
| | | |
Collapse
|
20
|
Hori S. Lineage stability and phenotypic plasticity of Foxp3+regulatory T cells. Immunol Rev 2014; 259:159-72. [DOI: 10.1111/imr.12175] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Shohei Hori
- Laboratory for Immune Homeostasis; RCAI; RIKEN Center for Integrative Medical Sciences; Kanagawa Japan
| |
Collapse
|
21
|
OHNO H. Gut microbiota, host defense and immunity: analysis with integrative omics approach. ACTA ACUST UNITED AC 2014; 37:403-11. [DOI: 10.2177/jsci.37.403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
22
|
Josefowicz SZ. Regulators of chromatin state and transcription in CD4 T-cell polarization. Immunology 2013; 139:299-308. [PMID: 23590627 DOI: 10.1111/imm.12115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/03/2013] [Indexed: 12/31/2022] Open
Abstract
Mature naive CD4 T-cells possess the potential for an array of highly specialized functions, from inflammatory to potently suppressive. This potential is encoded in regulatory DNA elements and is fulfilled through modification of chromatin and selective activation by the collaborative function of diverse transcription factors in response to environmental cues. The mechanisms and strategies employed by transcription factors for the programming of CD4 T-cell subsets will be discussed. In particular, the focus will be on co-operative activity of environmental response factors in the initial activation of regulatory DNA elements and chromatin alteration, and the subsequent role of 'master regulator' transcription factors in defining the fidelity and environmental responsiveness of different CD4 T-cell subsets.
Collapse
Affiliation(s)
- Steven Z Josefowicz
- Laboratory of Chromatin Biology and Epigenetics, Rockefeller University, New York, NY, USA.
| |
Collapse
|
23
|
Abstract
Regulatory T (TReg) cells are crucial for the prevention of fatal autoimmunity in mice and humans. Forkhead box P3 (FOXP3)(+) TReg cells are produced in the thymus and are also generated from conventional CD4(+) T cells in peripheral sites. It has been suggested that FOXP3(+) TReg cells might become unstable under certain inflammatory conditions and might adopt a phenotype that is more characteristic of effector CD4(+) T cells. These suggestions have caused considerable debate in the field and have important implications for the therapeutic use of TReg cells. In this article, Nature Reviews Immunology asks several experts for their views on the plasticity and stability of TReg cells.
Collapse
|
24
|
Knosp CA, Schiering C, Spence S, Carroll HP, Nel HJ, Osbourn M, Jackson R, Lyubomska O, Malissen B, Ingram R, Fitzgerald DC, Powrie F, Fallon PG, Johnston JA, Kissenpfennig A. Regulation of Foxp3+ inducible regulatory T cell stability by SOCS2. THE JOURNAL OF IMMUNOLOGY 2013; 190:3235-45. [PMID: 23455506 DOI: 10.4049/jimmunol.1201396] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Suppressor of cytokine signaling (SOCS) proteins are key regulators of CD4(+) T cell differentiation, and in particular, we have recently shown that SOCS2 inhibits the development of Th2 cells and allergic immune responses. Interestingly, transcriptome analyses have identified SOCS2 as being preferentially expressed in both natural regulatory T cells (Tregs) and inducible Tregs (iTregs); however, the role of SOCS2 in Foxp3(+) Treg function or development has not been fully elucidated. In this study, we show that despite having no effect on natural Treg development or function, SOCS2 is highly expressed in iTregs and required for the stable expression of Foxp3 in iTregs in vitro and in vivo. Indeed, SOCS2-deficient CD4(+) T cells upregulated Foxp3 following in vitro TGF-β stimulation, but failed to maintain stable expression of Foxp3. Moreover, in vivo generation of iTregs following OVA feeding was impaired in the absence of SOCS2 and could be rescued in the presence of IL-4 neutralizing Ab. Following IL-4 stimulation, SOCS2-deficient Foxp3(+) iTregs secreted elevated IFN-γ and IL-13 levels and displayed enhanced STAT6 phosphorylation. Therefore, we propose that SOCS2 regulates iTreg stability by downregulating IL-4 signaling. Moreover, SOCS2 is essential to maintain the anti-inflammatory phenotype of iTregs by preventing the secretion of proinflammatory cytokines. Collectively, these results suggest that SOCS2 may prevent IL-4-induced Foxp3(+) iTreg instability. Foxp3(+) iTregs are key regulators of immune responses at mucosal surfaces; therefore, this dual role of SOCS2 in both Th2 and Foxp3(+) iTregs reinforces SOCS2 as a potential therapeutic target for Th2-biased diseases.
Collapse
Affiliation(s)
- Camille A Knosp
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Belfast BT9 7AE, Northern Ireland, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Upregulation of CD200 is associated with Foxp3+ regulatory T cell expansion and disease progression in acute myeloid leukemia. Tumour Biol 2012. [DOI: 10.1007/s13277-012-0578-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|