101
|
Scheinecker C, Göschl L, Bonelli M. Treg cells in health and autoimmune diseases: New insights from single cell analysis. J Autoimmun 2019; 110:102376. [PMID: 31862128 DOI: 10.1016/j.jaut.2019.102376] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023]
Abstract
Autoimmune diseases, such as Systemic Lupus Erythematosus (SLE) or Rheumatoid Arthritis (RA) are characterized by the breakdown of immunological tolerance. Defects of regulatory T cells have been described among the various mechanisms, that are important for the development of autoimmune diseases, due to their critical role as regulators of peripheral immune tolerance and homeostasis. Initially T suppressor cells have been described as one population of peripheral T cells. Based on new technological advances a new understanding of the heterogeneity of different Treg cell populations in the lymphoid and non-lymphoid tissue has evolved over the last years. While initially Foxp3 has been defined as the main master regulator of Treg cells, we have learned that Treg cells from various tissue can be identified by a specific transcriptomic and epigenetic signature. Epigenetic mechanisms allow Treg cell stability, but we have also learned that certain Treg subsets are plastic and can under specific circumstances even enhance autoimmunity and inflammatory processes. Quantitative and functional defects of Treg cells have been observed in a variety of autoimmune diseases. Due to our understanding of the nature of this cell population, Treg cells have been a target of new Treg based therapies, such as low-dose IL-2. In addition, ongoing clinical trials aim to test safety and efficacy of transferred, in vitro expanded Treg cells in patients with autoimmune diseases and transplant patients.
Collapse
Affiliation(s)
- Clemens Scheinecker
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - Lisa Göschl
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - Michael Bonelli
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
102
|
Shi H, Chi H. Metabolic Control of Treg Cell Stability, Plasticity, and Tissue-Specific Heterogeneity. Front Immunol 2019; 10:2716. [PMID: 31921097 PMCID: PMC6917616 DOI: 10.3389/fimmu.2019.02716] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Regulatory T (Treg) cells are crucial for peripheral immune tolerance and prevention of autoimmunity and tissue damage. Treg cells are inherently defined by the expression of the transcription factor Foxp3, which enforces lineage development and immune suppressive function of these cells. Under various conditions as observed in autoimmunity, cancer and non-lymphoid tissues, a proportion of Treg cells respond to specific environmental signals and display altered stability, plasticity and tissue-specific heterogeneity, which further shape their context-dependent suppressive functions. Recent studies have revealed that metabolic programs play pivotal roles in controlling these processes in Treg cells, thereby considerably expanding our understanding of Treg cell biology. Here we summarize these recent advances that highlight how cell-extrinsic factors, such as nutrients, vitamins and metabolites, and cell-intrinsic metabolic programs, orchestrate Treg cell stability, plasticity, and tissue-specific heterogeneity. Understanding metabolic regulation of Treg cells should provide new insight into immune homeostasis and disease, with important therapeutic implications for autoimmunity, cancer, and other immune-mediated disorders.
Collapse
Affiliation(s)
- Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
103
|
Zhou H, Qin P, Liu Q, Yuan C, Hao Y, Zhang H, Wang Z, Ran X, Chu X, Yu W, Wang X, Hou Y, Peng J, Hou M. A prospective, multicenter study of low dose decitabine in adult patients with refractory immune thrombocytopenia. Am J Hematol 2019; 94:1374-1381. [PMID: 31591739 DOI: 10.1002/ajh.25646] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022]
Abstract
We conducted a prospective, multicenter study to evaluate the efficacy and safety of low-dose decitabine in adult patients with refractory immune thrombocytopenia. Adult patients who did not respond to, did not tolerate, or were unwilling to undergo splenectomy, with either a baseline platelet count less than 30 × 109 /L or the presence of bleeding symptoms and further need of ITP-specific treatments, were enrolled. Patients received decitabine at 3.5 mg/m2 intravenously for three consecutive days per cycle, for three cycles with a four-week interval between cycles. All patients were assessed every week during the first 12 weeks and at four-week intervals thereafter. We screened 49 patients for eligibility. Four patients were excluded and 45 received decitabine. At the end of decitabine treatment, complete response was achieved in eight patients (17.78%), and partial response was achieved in 15 patients (33.33%). The median time to initial response was 28 days (range, 14-70 days). Furthermore, seven relapsed patients received decitabine retreatment and all showed platelet response, including one complete response and six partial responses. Sustained response rates at 6, 12 and 18 months were 44.44% (20/45), 31.11% (14/45) and 20.0% (9/45), respectively. For responders, immune thrombocytopenia-related symptoms, fatigue, psychological health, fear, and overall quality of life were significantly improved. Adverse events were observed in 13 (28.89%) patients. No serious adverse events were recorded. In conclusion, low dose decitabine is potentially effective and safe in the management of adults with refractory immune thrombocytopenia. This trial is registered with clinicaltrials.gov identifier: NCT01568333.
Collapse
Affiliation(s)
- Hai Zhou
- Department of Hematology Qilu Hospital, Shandong University Jinan China
| | - Ping Qin
- Department of Hematology Qilu Hospital, Shandong University Jinan China
| | - Qiang Liu
- Department of Hematology Qilu Hospital, Shandong University Jinan China
| | - Chenglu Yuan
- Department of Hematology Qilu Hospital (Qingdao), Shandong University Qingdao China
| | - Yunliang Hao
- Department of Hematology Jining No.1 People's Hospital Jining China
| | - Haiyan Zhang
- Department of Hematology Linyi People's Hospital Linyi China
| | - Zhencheng Wang
- Department of Hematology Zibo Central Hospital Zibo China
| | - Xuehong Ran
- Department of Hematology Weifang People's Hospital Weifang China
| | - Xiaoxia Chu
- Department of Hematology Yantai Yuhuangding Hospital Yantai China
| | - Wenzheng Yu
- Department of Hematology Binzhou Medical University Hospital Binzhou China
| | - Xin Wang
- Department of Hematology Provincial Hospital Affiliated to Shandong University Jinan China
| | - Yu Hou
- Department of Hematology Qilu Hospital, Shandong University Jinan China
| | - Jun Peng
- Department of Hematology Qilu Hospital, Shandong University Jinan China
- Shandong Provincial Key Laboratory of Immunohematology Qilu Hospital, Shandong University Jinan China
- Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province Qilu Hospital, Shandong University Jinan China
| | - Ming Hou
- Department of Hematology Qilu Hospital, Shandong University Jinan China
- Shandong Provincial Key Laboratory of Immunohematology Qilu Hospital, Shandong University Jinan China
- Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province Qilu Hospital, Shandong University Jinan China
| |
Collapse
|
104
|
Jia X, Yang W, Zhou X, Han L, Shi J. Influence of demethylation on regulatory T and Th17 cells in myelodysplastic syndrome. Oncol Lett 2019; 19:442-448. [PMID: 31897157 PMCID: PMC6924080 DOI: 10.3892/ol.2019.11114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndrome (MDS) represents a heterogeneous hematopoietic disorder in which mature blood cells are derived from an abnormal multipotent progenitor cell. The current therapy for MDS involves repeated cycles of DNA methyltransferase (DNMT) inhibitors, particularly the demethylation drug 5-azacytidine (5-azaC) which has been shown to increase the survival of patients with high-risk MDS. The mechanisms behind the therapeutic effects of 5-azaC are not yet clear. In this study the effect of 5-azaC on the development of regulatory T cells (Tregs) and T-helper 17 (Th17) cells was investigated. The numbers of CD4+ T-cell subsets in 30 patients with intermediate-2/high-risk MDS were serially assessed at diagnosis and following 5-azaC treatment. The number of FoxP3+ Tregs was significantly higher after 3 months of therapy. However, there was no statistical difference in the number of Th17 cells following treatment. In vitro, 5-azaC enhanced the overall proportion of Tregs, but not Th17, in CD4+ T cells from patients with MDS. Addition of 5-azaC reduced the proliferative capacity of Tregs, suggesting that the increase in Tregs was due to conversion of conventional CD25− cells, rather than proliferation of CD25+FoxP3+ cells. The FoxP3 expression in 5-azaC-treated T effectors was also increased. Interestingly, while Tbet and RORγT mRNA transcription had no obvious changes, due to the demethylation of the FoxP3 promoter, these findings are important in associating the induction of DNA hypomethylation and the clinical response to 5-azaC.
Collapse
Affiliation(s)
- Xinyan Jia
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Wenzhong Yang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Xiaohui Zhou
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Lu Han
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Jumei Shi
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
105
|
Hudon Thibeault AA, Laprise C. Cell-Specific DNA Methylation Signatures in Asthma. Genes (Basel) 2019; 10:E932. [PMID: 31731604 PMCID: PMC6896152 DOI: 10.3390/genes10110932] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022] Open
Abstract
Asthma is a complex trait, often associated with atopy. The genetic contribution has been evidenced by familial occurrence. Genome-wide association studies allowed for associating numerous genes with asthma, as well as identifying new loci that have a minor contribution to its phenotype. Considering the role of environmental exposure on asthma development, an increasing amount of literature has been published on epigenetic modifications associated with this pathology and especially on DNA methylation, in an attempt to better understand its missing heritability. These studies have been conducted in different tissues, but mainly in blood or its peripheral mononuclear cells. However, there is growing evidence that epigenetic changes that occur in one cell type cannot be directly translated into another one. In this review, we compare alterations in DNA methylation from different cells of the immune system and of the respiratory tract. The cell types in which data are obtained influences the global status of alteration of DNA methylation in asthmatic individuals compared to control (an increased or a decreased DNA methylation). Given that several genes were cell-type-specific, there is a great need for comparative studies on DNA methylation from different cells, but from the same individuals in order to better understand the role of epigenetics in asthma pathophysiology.
Collapse
Affiliation(s)
- Andrée-Anne Hudon Thibeault
- Département des sciences fondamentales, Université du Québec à Chicoutimi (UQAC), Saguenay, G7H 2B1 QC, Canada;
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi (UQAC), Saguenay, G7H 2B1 QC, Canada
- Quebec Respiratory Health Network, Quebec, G1V 4G5 QC, Canada
| | - Catherine Laprise
- Département des sciences fondamentales, Université du Québec à Chicoutimi (UQAC), Saguenay, G7H 2B1 QC, Canada;
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi (UQAC), Saguenay, G7H 2B1 QC, Canada
- Quebec Respiratory Health Network, Quebec, G1V 4G5 QC, Canada
| |
Collapse
|
106
|
Epigenetic mechanisms regulating T-cell responses. J Allergy Clin Immunol 2019; 142:728-743. [PMID: 30195378 DOI: 10.1016/j.jaci.2018.07.014] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022]
Abstract
During the last decade, advances in sequencing technologies allowed production of a wealth of information on epigenetic modifications in T cells. Epigenome maps, in combination with mechanistic studies, have demonstrated that T cells undergo extensive epigenome remodeling in response to signals, which has a strong effect on phenotypic stability and function of lymphocytes. In this review we focus on DNA methylation, histone modifications, and chromatin structure as important epigenetic mechanisms involved in controlling T-cell responses. In particular, we discuss epigenetic processes in light of the development, activation, and differentiation of CD4+ T helper (TH), regulatory T, and CD8+ T cells. As central aspects of the adaptive immune system, we review mechanisms that ensure molecular memory, stability, plasticity, and exhaustion of T cells. We further discuss the effect of the tissue environment on imprinting T-cell epigenomes with potential implications for immunotherapy.
Collapse
|
107
|
Gambacorta V, Gnani D, Vago L, Di Micco R. Epigenetic Therapies for Acute Myeloid Leukemia and Their Immune-Related Effects. Front Cell Dev Biol 2019; 7:207. [PMID: 31681756 PMCID: PMC6797914 DOI: 10.3389/fcell.2019.00207] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/11/2019] [Indexed: 12/19/2022] Open
Abstract
Over the past decades, our molecular understanding of acute myeloid leukemia (AML) pathogenesis dramatically increased, thanks also to the advent of next-generation sequencing (NGS) technologies. Many of these findings, however, have not yet translated into new prognostic markers or rationales for treatments. We now know that AML is a highly heterogeneous disease characterized by a very low mutational burden. Interestingly, the few mutations identified mainly reside in epigenetic regulators, which shape and define leukemic cell identity. In the light of these discoveries and given the increasing number of drugs targeting epigenetic regulators in clinical development and testing, great interest is emerging for the use of small molecules targeting leukemia epigenome. Together with their effects on leukemia cell-intrinsic properties, such as proliferation and survival, epigenetic drugs may affect the way leukemic cells communicate with the surrounding components of the tumor and immune microenvironment. Here, we review current knowledge on alterations in the AML epigenetic landscape and discuss the promises of epigenetic therapies for AML treatment. Finally, we summarize emerging molecular studies elucidating how epigenetic rewiring in cancer cells may as well exert immune-modulatory functions, boost the immune system, and potentially contribute to better patient outcomes.
Collapse
Affiliation(s)
- Valentina Gambacorta
- Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy.,Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Milano-Bicocca University, Milan, Italy
| | - Daniela Gnani
- Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Vago
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Di Micco
- Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
108
|
Herppich S, Toker A, Pietzsch B, Kitagawa Y, Ohkura N, Miyao T, Floess S, Hori S, Sakaguchi S, Huehn J. Dynamic Imprinting of the Treg Cell-Specific Epigenetic Signature in Developing Thymic Regulatory T Cells. Front Immunol 2019; 10:2382. [PMID: 31681278 PMCID: PMC6797672 DOI: 10.3389/fimmu.2019.02382] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022] Open
Abstract
Regulatory T (Treg) cells mainly develop within the thymus and arise from CD25+Foxp3− (CD25+ TregP) or CD25−Foxp3+ (Foxp3+ TregP) Treg cell precursors resulting in Treg cells harboring distinct transcriptomic profiles and complementary T cell receptor repertoires. The stable and long-term expression of Foxp3 in Treg cells and their stable suppressive phenotype are controlled by the demethylation of Treg cell-specific epigenetic signature genes including an evolutionarily conserved CpG-rich element within the Foxp3 locus, the Treg-specific demethylated region (TSDR). Here we analyzed the dynamics of the imprinting of the Treg cell-specific epigenetic signature genes in thymic Treg cells. We could demonstrate that CD25+Foxp3+ Treg cells show a progressive demethylation of most signature genes during maturation within the thymus. Interestingly, a partial demethylation of several Treg cell-specific epigenetic signature genes was already observed in Foxp3+ TregP but not in CD25+ TregP. Furthermore, Foxp3+ TregP were very transient in nature and arose at a more mature developmental stage when compared to CD25+ TregP. When the two Treg cell precursors were cultured in presence of IL-2, a factor known to be critical for thymic Treg cell development, we observed a major impact of IL-2 on the demethylation of the TSDR with a more pronounced effect on Foxp3+ TregP. Together, these results suggest that the establishment of the Treg cell-specific hypomethylation pattern is a continuous process throughout thymic Treg cell development and that the two known Treg cell precursors display distinct dynamics for the imprinting of the Treg cell-specific epigenetic signature genes.
Collapse
Affiliation(s)
- Susanne Herppich
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Aras Toker
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Beate Pietzsch
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Yohko Kitagawa
- Laboratory of Experimental Immunology, World Premier International Research Center Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Naganari Ohkura
- Laboratory of Experimental Immunology, World Premier International Research Center Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takahisa Miyao
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Shohei Hori
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Laboratory of Immunology and Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, World Premier International Research Center Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
109
|
Ramos PS. Epigenetics of scleroderma: Integrating genetic, ethnic, age, and environmental effects. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2019; 4:238-250. [PMID: 35382507 PMCID: PMC8922566 DOI: 10.1177/2397198319855872] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/15/2019] [Indexed: 08/02/2023]
Abstract
Scleroderma or systemic sclerosis is thought to result from the interplay between environmental or non-genetic factors in a genetically susceptible individual. Epigenetic modifications are influenced by genetic variation and environmental exposures, and change with chronological age and between populations. Despite progress in identifying genetic, epigenetic, and environmental risk factors, the underlying mechanism of systemic sclerosis remains unclear. Since epigenetics provides the regulatory mechanism linking genetic and non-genetic factors to gene expression, understanding the role of epigenetic regulation in systemic sclerosis will elucidate how these factors interact to cause systemic sclerosis. Among the cell types under tight epigenetic control and susceptible to epigenetic dysregulation, immune cells are critically involved in early pathogenic events in the progression of fibrosis and systemic sclerosis. This review starts by summarizing the changes in DNA methylation, histone modification, and non-coding RNAs associated with systemic sclerosis. It then discusses the role of genetic, ethnic, age, and environmental effects on epigenetic regulation, with a focus on immune system dysregulation. Given the potential of epigenome editing technologies for cell reprogramming and as a therapeutic approach for durable gene regulation, this review concludes with a prospect on epigenetic editing. Although epigenomics in systemic sclerosis is in its infancy, future studies will help elucidate the regulatory mechanisms underpinning systemic sclerosis and inform the design of targeted epigenetic therapies to control its dysregulation.
Collapse
Affiliation(s)
- Paula S Ramos
- Paula S. Ramos, Division of Rheumatology and Immunology, Department of Medicine and Department of Public Health Sciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 816, MSC 637, Charleston, SC 29425, USA.
| |
Collapse
|
110
|
Zhang Z, Zhou X. Foxp3 Instability Helps tTregs Distinguish Self and Non-self. Front Immunol 2019; 10:2226. [PMID: 31608056 PMCID: PMC6769115 DOI: 10.3389/fimmu.2019.02226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/03/2019] [Indexed: 12/02/2022] Open
Abstract
Regulatory T cells (Tregs) are small subsets of CD4 T cells that play a central role in the controlling of immune tolerance. Tregs are either generated in the thymus (tTregs) or the periphery (pTregs), and both express the master transcription factor Foxp3. Stable expression of Foxp3 is important for the maintenance of Tregs identity and their suppressive function. Similar to conventional T cells, Tregs can recognize both self- and non-self-antigens, and TCR engagement leads to Treg activation and the generation of effector Tregs. Emerging shreds of evidence suggest Tregs are not always stable, even fully committed mature tTregs, and can lose foxp3 expression and programming to effector-like T cells. In this review, we summarize recent findings in Treg instability and the intrinsic and extrinsic mechanisms in controlling the Foxp3 expression. Finally, we propose a new hypothesis that Foxp3 instability might help tTregs distinguish between self and non-self-antigens.
Collapse
Affiliation(s)
- Zhongmei Zhang
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xuyu Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
111
|
Sun X, Cui Y, Feng H, Liu H, Liu X. TGF-β signaling controls Foxp3 methylation and T reg cell differentiation by modulating Uhrf1 activity. J Exp Med 2019; 216:2819-2837. [PMID: 31515281 PMCID: PMC6888975 DOI: 10.1084/jem.20190550] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/28/2019] [Accepted: 08/28/2019] [Indexed: 01/02/2023] Open
Abstract
Regulatory T (T reg) cells are required for the maintenance of immune homeostasis. Both TGF-β signaling and epigenetic modifications are important for Foxp3 induction, but how TGF-β signaling participates in the epigenetic regulation of Foxp3 remains largely unknown. Here we showed that T cell-specific ablation of Uhrf1 resulted in T reg-biased differentiation in TCR-stimulated naive T cells in the absence of TGF-β signaling, and these Foxp3+ T cells had a suppressive function. Adoptive transfer of Uhrf1 -/- naive T cells could significantly suppress colitis due to increased iT reg cell generation. Mechanistically, Uhrf1 was induced upon TCR stimulation and participated in the maintenance of DNA methylation patterns of T reg cell-specific genes during cell division, while it was phosphorylated upon TGF-β stimulation and sequestered outside the nucleus, and ultimately underwent proteasome-dependent degradation. Collectively, our study reveals a novel epigenetic mechanism of TGF-β-mediated iT reg cell differentiation by modulating Uhrf1 activity and suggests that Uhrf1 may be a potential therapeutic target in inflammatory diseases for generating stable iT reg cells.
Collapse
Affiliation(s)
- Xiang Sun
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yu Cui
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Haiyun Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Haifeng Liu
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiaolong Liu
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
112
|
Kanamori M, Nakatsukasa H, Ito M, Chikuma S, Yoshimura A. Reprogramming of Th1 cells into regulatory T cells through rewiring of the metabolic status. Int Immunol 2019; 30:357-373. [PMID: 29982622 DOI: 10.1093/intimm/dxy043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/28/2018] [Indexed: 12/16/2022] Open
Abstract
T helper type 1 (Th1) cells form one of the most stable CD4 T-cell subsets, and direct conversion of fully differentiated Th1 to regulatory T (Treg) cells has been poorly investigated. Here, we established a culture method for inducing Foxp3 from Th1 cells of mice and humans. This is achieved simply by resting Th1 cells without T-cell receptor ligation before stimulation in the presence of transforming growth factor-beta (TGF-β). We named the resulting Th1-derived Foxp3+ cells Th1reg cells. Mouse Th1reg cells showed an inducible Treg-like phenotype and suppressive ability both in vitro and in vivo. Th1reg cells could also be induced from in vivo-developed mouse Th1 cells. Unexpectedly, the resting process enabled Foxp3 expression not through epigenetic changes at the locus, but through metabolic change resulting from reduced mammalian target of rapamycin complex 1 (mTORC1) activity. mTORC1 suppressed TGF-β-induced phosphorylation of Smad2/3 in Th1 cells, which was restored in rested cells. Our study warrants future research aiming at development of immunotherapy with Th1reg cells.
Collapse
Affiliation(s)
- Mitsuhiro Kanamori
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroko Nakatsukasa
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Minako Ito
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
113
|
Karl M, Sommer C, Gabriel CH, Hecklau K, Venzke M, Hennig AF, Radbruch A, Selbach M, Baumgrass R. Recruitment of Histone Methyltransferase Ehmt1 to Foxp3 TSDR Counteracts Differentiation of Induced Regulatory T Cells. J Mol Biol 2019; 431:3606-3625. [PMID: 31362003 DOI: 10.1016/j.jmb.2019.07.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/13/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022]
Abstract
Differentiation toward CD4+ regulatory T (Treg) cells is essentially dependent on an epigenetic program at Treg signature genes, which involves remodeling of the Treg-specific demethylated regions (TSDRs). In particular, the epigenetic status of the conserved non-coding sequence 2 of Foxp3 (Foxp3 TSDR) determines expression stability of the master transcription factor and thus Treg lineage identity. However, the molecular mechanisms controlling the epigenetic remodeling at TSDRs in Treg and conventional T cells are largely unknown. Using a combined approach of DNA pull-down and mass spectrometric analysis, we report a novel regulatory mechanism in which transcription factor Wiz recruits the histone methyltransferase Ehmt1 to Foxp3 TSDR. We show that both Wiz and Ehmt1 are crucial for shaping the region with the repressive histone modification H3K9me2 in conventional T cells. Consistently, knocking out either Ehmt1 or Wiz by CRISPR/Cas resulted in the loss of H3K9me2 and enhanced Foxp3 expression during iTreg differentiation. Moreover, the essential role of the Wiz-Ehmt1 interaction as observed at several TSDRs indicates a global function of Ehmt1 in the Treg differentiation program.
Collapse
Affiliation(s)
- Martin Karl
- Signal Transduction, German Rheumatism Research Center (DRFZ), A Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Christian Sommer
- Proteome Dynamics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Christian H Gabriel
- Signal Transduction, German Rheumatism Research Center (DRFZ), A Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Katharina Hecklau
- Signal Transduction, German Rheumatism Research Center (DRFZ), A Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Melanie Venzke
- Signal Transduction, German Rheumatism Research Center (DRFZ), A Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Anna Floriane Hennig
- Signal Transduction, German Rheumatism Research Center (DRFZ), A Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Andreas Radbruch
- Cell Biology, German Rheumatism Research Center (DRFZ), A Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany; Charité-University Medicine, Charitéplatz 1, 10117 Berlin, Germany
| | - Matthias Selbach
- Proteome Dynamics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Ria Baumgrass
- Signal Transduction, German Rheumatism Research Center (DRFZ), A Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
114
|
Park MJ, Moon SJ, Lee EJ, Kim EK, Baek JA, Kim SY, Jung KA, Lee SH, Choi JW, Kim DS, Min JK, Park SH, Shin D, Cho ML. Daurinol Attenuates Autoimmune Arthritis via Stabilization of Nrp1-PTEN-Foxp3 Signaling in Regulatory T Cells. Front Immunol 2019; 10:1526. [PMID: 31379809 PMCID: PMC6651269 DOI: 10.3389/fimmu.2019.01526] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 06/18/2019] [Indexed: 12/23/2022] Open
Abstract
Optimizing Treg function and improving Treg stability are attractive treatment strategies for treating autoimmune rheumatoid arthritis (RA). However, the limited number of circulating Tregs and questions about the functional stability of in vitro-expanded Tregs are potential limitations of Treg-based cell therapy. The aim of this study was to analyze the regulatory effect of daurinol, a catalytic inhibitor of topoisomerase IIα, on Th cell differentiation and to evaluate their therapeutic potential in a preclinical experimental model of RA. We investigated the effect of daurinol on T cell differentiation by flow cytometry. Foxp3 stability and methylation were analyzed by suppression assays and bisulfite pyrosequencing. Daurinol was treated in the collagen-induced arthritis (CIA) model, and the effects in vivo were determined. We found that daurinol can promote Treg differentiation and reciprocally inhibit Th17 differentiation. This Treg-inducing property of daurinol was associated with decreased activity of Akt-mTOR and reciprocally increased activity of neuropilin-1 (Nrp1)-PTEN. Daurinol treatment inhibited aerobic glycolysis in Th17 conditions, indicating the metabolic changes by daurinol. We found that the daurinol increase the Treg stability was achieved by Foxp3 hypomethylation. In vivo daurinol treatment in CIA mice reduced the clinical arthritis severity and histological inflammation. The Treg population frequency increased and the Th17 cells decreased in the spleens of arthritis mice treated with daurinol. These results showed the anti-arthritic and immunoregulating properties of daurinol is achieved by increased differentiation and stabilization of Tregs. Our study provides first evidence for daurinol as a treatment for RA.
Collapse
Affiliation(s)
- Min-Jung Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Su-Jin Moon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Jung Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jin-Ah Baek
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Se-Young Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Kyung Ah Jung
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung Hoon Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jeong Won Choi
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Da-Som Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jun-Ki Min
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Dongyun Shin
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
115
|
Moldenhauer LM, Schjenken JE, Hope CM, Green ES, Zhang B, Eldi P, Hayball JD, Barry SC, Robertson SA. Thymus-Derived Regulatory T Cells Exhibit Foxp3 Epigenetic Modification and Phenotype Attenuation after Mating in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 203:647-657. [DOI: 10.4049/jimmunol.1900084] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/20/2019] [Indexed: 12/30/2022]
|
116
|
Vigano S, Alatzoglou D, Irving M, Ménétrier-Caux C, Caux C, Romero P, Coukos G. Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function. Front Immunol 2019; 10:925. [PMID: 31244820 PMCID: PMC6562565 DOI: 10.3389/fimmu.2019.00925] [Citation(s) in RCA: 269] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
T cells play a critical role in cancer control, but a range of potent immunosuppressive mechanisms can be upregulated in the tumor microenvironment (TME) to abrogate their activity. While various immunotherapies (IMTs) aiming at re-invigorating the T-cell-mediated anti-tumor response, such as immune checkpoint blockade (ICB), and the adoptive cell transfer (ACT) of natural or gene-engineered ex vivo expanded tumor-specific T cells, have led to unprecedented clinical responses, only a small proportion of cancer patients benefit from these treatments. Important research efforts are thus underway to identify biomarkers of response, as well as to develop personalized combinatorial approaches that can target other inhibitory mechanisms at play in the TME. In recent years, adenosinergic signaling has emerged as a powerful immuno-metabolic checkpoint in tumors. Like several other barriers in the TME, such as the PD-1/PDL-1 axis, CTLA-4, and indoleamine 2,3-dioxygenase (IDO-1), adenosine plays important physiologic roles, but has been co-opted by tumors to promote their growth and impair immunity. Several agents counteracting the adenosine axis have been developed, and pre-clinical studies have demonstrated important anti-tumor activity, alone and in combination with other IMTs including ICB and ACT. Here we review the regulation of adenosine levels and mechanisms by which it promotes tumor growth and broadly suppresses protective immunity, with extra focus on the attenuation of T cell function. Finally, we present an overview of promising pre-clinical and clinical approaches being explored for blocking the adenosine axis for enhanced control of solid tumors.
Collapse
Affiliation(s)
- Selena Vigano
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dimitrios Alatzoglou
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christine Ménétrier-Caux
- Department of Immunology Virology and Inflammation, INSERM 1052, CNRS 5286, Léon Bérard Cancer Center, Cancer Research Center of Lyon, University of Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Christophe Caux
- Department of Immunology Virology and Inflammation, INSERM 1052, CNRS 5286, Léon Bérard Cancer Center, Cancer Research Center of Lyon, University of Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Pedro Romero
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
117
|
Foxp1 is critical for the maintenance of regulatory T-cell homeostasis and suppressive function. PLoS Biol 2019; 17:e3000270. [PMID: 31125332 PMCID: PMC6534289 DOI: 10.1371/journal.pbio.3000270] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 04/30/2019] [Indexed: 12/13/2022] Open
Abstract
Regulatory T (Treg) cells play central roles in maintaining immune homeostasis and self-tolerance. However, the molecular mechanisms underlying Treg cell homeostasis and suppressive function are still not fully understood. Here, we report that the deletion of another P subfamily members of the forkhead box (Foxp) subfamily member Foxp1 in Treg cells led to increased numbers of activated Treg (aTreg) cells at the expense of quiescent Treg cells, and also resulted in impaired Treg suppressive function. Mice with Foxp1-deficient Treg cells developed spontaneous inflammatory disease with age; they also had more severe inflammatory disease in colitis and experimental autoimmune encephalomyelitis (EAE) models. Mechanistically, we found that Foxp1 bound to the conserved noncoding sequence 2 (CNS2) element of the Foxp3 locus and helped maintain Treg suppressive function by stabilizing the Foxp3 expression. Furthermore, we found that Foxp1 and Foxp3 coordinated the regulation of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) expression levels. Taken together, our study demonstrates that Foxp1 plays critical roles in both maintaining Treg cell quiescence during homeostasis and regulating Treg suppressive function. The transcription factor Foxp3 has been considered as the master regulator of regulatory T cells (Tregs), but this study shows that another Foxp subfamily member, Foxp1, plays important roles in the homeostasis, stability, and suppressor function of Tregs.
Collapse
|
118
|
Bartolotti N, Lazarov O. CREB signals as PBMC-based biomarkers of cognitive dysfunction: A novel perspective of the brain-immune axis. Brain Behav Immun 2019; 78:9-20. [PMID: 30641141 PMCID: PMC6488430 DOI: 10.1016/j.bbi.2019.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/21/2018] [Accepted: 01/10/2019] [Indexed: 12/26/2022] Open
Abstract
To date, there is no reliable biomarker for the assessment or determination of cognitive dysfunction in Alzheimer's disease and related dementia. Such a biomarker would not only aid in diagnostics, but could also serve as a measure of therapeutic efficacy. It is widely acknowledged that the hallmarks of Alzheimer's disease, namely, amyloid deposits and neurofibrillary tangles, as well as their precursors and metabolites, are poorly correlated with cognitive function and disease stage and thus have low diagnostic or prognostic value. A lack of biomarkers is one of the major roadblocks in diagnosing the disease and in assessing the efficacy of potential therapies. The phosphorylation of cAMP Response Element Binding protein (pCREB) plays a major role in memory acquisition and consolidation. In the brain, CREB activation by phosphorylation at Ser133 and the recruitment of transcription cofactors such as CREB binding protein (CBP) is a critical step for the formation of memory. This set of processes is a prerequisite for the transcription of genes thought to be important for synaptic plasticity, such as Egr-1. Interestingly, recent work suggests that the expression of pCREB in peripheral blood mononuclear cells (PBMC) positively correlates with pCREB expression in the postmortem brain of Alzheimer's patients, suggesting not only that pCREB expression in PBMC might serve as a biomarker of cognitive dysfunction, but also that the dysfunction of CREB signaling may not be limited to the brain in AD, and that a link may exist between the regulation of CREB in the blood and in the brain. In this review we consider the evidence suggesting a correlation between the level of CREB signals in the brain and blood, the current knowledge about CREB in PBMC and its association with CREB in the brain, and the implications and mechanisms for a neuro-immune cross talk that may underlie this communication. This Review will discuss the possibility that peripheral dysregulation of CREB is an early event in AD pathogenesis, perhaps as a facet of immune system dysfunction, and that this impairment in peripheral CREB signaling modifies CREB signaling in the brain, thus exacerbating cognitive decline in AD. A more thorough understanding of systemic dysregulation of CREB in AD will facilitate the search for a biomarker of cognitive function in AD, and also aid in the understanding of the mechanisms underlying cognitive decline in AD.
Collapse
Affiliation(s)
- Nancy Bartolotti
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
119
|
Göschl L, Scheinecker C, Bonelli M. Treg cells in autoimmunity: from identification to Treg-based therapies. Semin Immunopathol 2019; 41:301-314. [PMID: 30953162 DOI: 10.1007/s00281-019-00741-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/22/2019] [Indexed: 12/27/2022]
Abstract
Regulatory (Treg) cells are key regulators of inflammation and important for immune tolerance and homeostasis. A major progress has been made in the identification and classification of Treg cells. Due to technological advances, we have gained deep insights in the epigenetic regulation of Treg cells. The use of fate reporter mice allowed addressing the functional consequences of loss of Foxp3 expression. Depending on the environment Treg cells gain effector functions upon loss of Foxp3 expression. However, the traditional view that Treg cells become necessarily pathogenic by gaining effector functions was challenged by recent findings and supports the notion of Treg cell lineage plasticity. Treg cell stability is also a major issue for Treg cell therapies. Clinical trials are designed to use polyclonal Treg cells as therapeutic tools. Here, we summarize the role of Treg cells in selected autoimmune diseases and recent advances in the field of Treg targeted therapies.
Collapse
Affiliation(s)
- Lisa Göschl
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Clemens Scheinecker
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Michael Bonelli
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
120
|
Attias M, Al-Aubodah T, Piccirillo CA. Mechanisms of human FoxP3 + T reg cell development and function in health and disease. Clin Exp Immunol 2019; 197:36-51. [PMID: 30864147 PMCID: PMC6591147 DOI: 10.1111/cei.13290] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2019] [Indexed: 12/18/2022] Open
Abstract
Regulatory T (Treg) cells represent an essential component of peripheral tolerance. Given their potently immunosuppressive functions that is orchestrated by the lineage‐defining transcription factor forkhead box protein 3 (FoxP3), clinical modulation of these cells in autoimmunity and cancer is a promising therapeutic target. However, recent evidence in mice and humans indicates that Treg cells represent a phenotypically and functionally heterogeneic population. Indeed, both suppressive and non‐suppressive Treg cells exist in human blood that are otherwise indistinguishable from one another using classical Treg cell markers such as CD25 and FoxP3. Moreover, murine Treg cells display a degree of plasticity through which they acquire the trafficking pathways needed to home to tissues containing target effector T (Teff) cells. However, this plasticity can also result in Treg cell lineage instability and acquisition of proinflammatory Teff cell functions. Consequently, these dysfunctional CD4+FoxP3+ T cells in human and mouse may fail to maintain peripheral tolerance and instead support immunopathology. The mechanisms driving human Treg cell dysfunction are largely undefined, and obscured by the scarcity of reliable immunophenotypical markers and the disregard paid to Treg cell antigen‐specificity in functional assays. Here, we review the mechanisms controlling the stability of the FoxP3+ Treg cell lineage phenotype. Particular attention will be paid to the developmental and functional heterogeneity of human Treg cells, and how abrogating these mechanisms can lead to lineage instability and Treg cell dysfunction in diseases like immunodysregulation polyendocrinopathy enteropathy X‐linked (IPEX) syndrome, type 1 diabetes, rheumatoid arthritis and cancer.
Collapse
Affiliation(s)
- M Attias
- Program in Infectious Diseases and Immunology in Global Health, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada.,Centre of Excellence in Translational Immunology (CETI), Montréal, Québec, Canada
| | - T Al-Aubodah
- Program in Infectious Diseases and Immunology in Global Health, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada.,Centre of Excellence in Translational Immunology (CETI), Montréal, Québec, Canada
| | - C A Piccirillo
- Program in Infectious Diseases and Immunology in Global Health, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada.,Centre of Excellence in Translational Immunology (CETI), Montréal, Québec, Canada
| |
Collapse
|
121
|
Morales-Nebreda L, McLafferty FS, Singer BD. DNA methylation as a transcriptional regulator of the immune system. Transl Res 2019; 204:1-18. [PMID: 30170004 PMCID: PMC6331288 DOI: 10.1016/j.trsl.2018.08.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
DNA methylation is a dynamic epigenetic modification with a prominent role in determining mammalian cell development, lineage identity, and transcriptional regulation. Primarily linked to gene silencing, novel technologies have expanded the ability to measure DNA methylation on a genome-wide scale and uncover context-dependent regulatory roles. The immune system is a prototypic model for studying how DNA methylation patterning modulates cell type- and stimulus-specific transcriptional programs. Preservation of host defense and organ homeostasis depends on fine-tuned epigenetic mechanisms controlling myeloid and lymphoid cell differentiation and function, which shape innate and adaptive immune responses. Dysregulation of these processes can lead to human immune system pathology as seen in blood malignancies, infections, and autoimmune diseases. Identification of distinct epigenotypes linked to pathogenesis carries the potential to validate therapeutic targets in disease prevention and management.
Collapse
Affiliation(s)
- Luisa Morales-Nebreda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Fred S McLafferty
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Benjamin D Singer
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
122
|
Khosravi M, Bidmeshkipour A, Moravej A, Hojjat-Assari S, Naserian S, Karimi MH. Induction of CD4 +CD25 +Foxp3 + regulatory T cells by mesenchymal stem cells is associated with RUNX complex factors. Immunol Res 2019; 66:207-218. [PMID: 29143918 DOI: 10.1007/s12026-017-8973-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Among the particular immunomodulation properties of mesenchymal stem cells (MSCs), one relies on their capacity to regulatory T cell (Treg) induction from effector T cells. Stable expression of Foxp3 has a dominant role in suppressive phenotype and stability of induced regulatory T cells (iTregs). How MSCs induce stable Foxp3 expression in iTregs remains unknown. We previously showed MSCs could enhance demethylation of Treg-specific demethylated region (TSDR) in iTregs in cell-cell contact manner (unpublished data). Here, we evaluated the possible effect of MSCs on the mRNA expression of Runx complex genes (Runx1, Runx3, and CBFB) that perch on TSDR in iTregs and play the main role in suppressive properties of Tregs, a regulatory pathway that has not yet been explored by MSCs. Also, we investigated the mRNA expression of MBD2 that promotes TSDR demethylation in Tregs. We first showed that in vitro MSC-iTreg induction was associated with strong mRNA modifications of genes involved in Runx complex. We next injected high doses of MSCs in a murine model of C57BL/6 into Balb/C allogeneic skin transplantation to prolong allograft survival. When splenocytes of grafted mice were analyzed, we realized that the Foxp3 expression was increased at day 5 and 10 post-graft merely in MSC-treated mice. Furthermore, Foxp3 mRNA expression was associated with modified Runx complex mRNA expression comparable to what was shown in in vitro studies. Hence, our data identify a possible mechanism in which MSCs convert conventional T cells to iTreg through strong modifications of mRNA of genes that are involved in Runx complex of Foxp3.
Collapse
Affiliation(s)
- Maryam Khosravi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.,Institut Français de Recherche et d'Enseignement Supérieur à l'International (IFRES-INT), Paris, France
| | - Ali Bidmeshkipour
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.
| | - Ali Moravej
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Suzzan Hojjat-Assari
- Institut Français de Recherche et d'Enseignement Supérieur à l'International (IFRES-INT), Paris, France
| | - Sina Naserian
- Inserm, U1197, Hôpital Paul Brousse, 94807, Villejuif, France
| | | |
Collapse
|
123
|
TSOKOS GEORGEC, TSOKOS MARIAG. TARGETING TARGETED TREATMENT FOR IMMUNE AND NON-IMMUNE KIDNEY DISEASES. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2019; 130:88-99. [PMID: 31516171 PMCID: PMC6735968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We have found that calcium calmodulin kinase IV is increased in T cells, podocytes, and mesangial cells from patients with systemic lupus erythematosus, as well as in lupus-prone mice, podocytes of patients with focal segmental glomerulosclerosis, and in mice injected with doxorubicin. We showed that this accounts for aberrant T cell function and glomerular damage. Using nanoparticles (nlg) loaded with a small drug inhibitor of calcium calmodulin kinase IV and tagged with antibodies directed to CD4 we have been able to show inhibition of autoimmunity and lupus nephritis. Also, using nlg tagged with antibodies to nephrin, we showed suppression of nephritis in lupus-prone mice and of glomerular damage in mice exposed to doxorubicin. We propose the development of approaches to deliver drugs to cells in a targeted and precise manner.
Collapse
Affiliation(s)
- GEORGE C. TSOKOS
- Correspondence and reprint requests: George C. Tsokos, MD, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School,
330 Brookline Ave., CLS 937, Boston, Massachusetts 02215617-735-4161
| | | |
Collapse
|
124
|
Silva Morales M, Mueller D. Anergy into T regulatory cells: an integration of metabolic cues and epigenetic changes at the Foxp3 conserved non-coding sequence 2. F1000Res 2018; 7. [PMID: 30613389 PMCID: PMC6305231 DOI: 10.12688/f1000research.16551.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2018] [Indexed: 01/12/2023] Open
Abstract
Peripheral immune self-tolerance relies on protective mechanisms to control autoreactive T cells that escape deletion in the thymus. Suppression of autoreactive lymphocytes is necessary to avoid autoimmunity and immune cell–mediated damage of healthy tissues. An intriguing relationship has emerged between two mechanisms of peripheral tolerance—induction of anergy and Foxp3
+ regulatory T (Treg) cells—and is not yet well understood. A subpopulation of autoreactive anergic CD4 T cells is a precursor of Treg cells. We now hypothesize that phenotypic and mechanistic features of Treg cells can provide insights to understand the mechanisms behind anergy-derived Treg cell differentiation. In this short review, we will highlight several inherent similarities between the anergic state in conventional CD4 T cells as compared with fully differentiated natural Foxp3
+ Treg cells and then propose a model whereby modulations in metabolic programming lead to changes in DNA methylation at the Foxp3 locus to allow
Foxp3 expression following the reversal of anergy.
Collapse
Affiliation(s)
- Milagros Silva Morales
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, and the University of Minnesota Medical School, Minneapolis, USA
| | - Daniel Mueller
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, and the University of Minnesota Medical School, Minneapolis, USA
| |
Collapse
|
125
|
Minskaia E, Saraiva BC, Soares MMV, Azevedo RI, Ribeiro RM, Kumar SD, Vieira AIS, Lacerda JF. Molecular Markers Distinguishing T Cell Subtypes With TSDR Strand-Bias Methylation. Front Immunol 2018; 9:2540. [PMID: 30455694 PMCID: PMC6230625 DOI: 10.3389/fimmu.2018.02540] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/15/2018] [Indexed: 12/28/2022] Open
Abstract
Human regulatory CD4+CD25+FOXP3+ T cells (Treg) play important roles in the maintenance of self-tolerance and immune homeostasis in various disease settings and are also involved in the suppression of effective immune responses. These cells are heterogeneous in phenotype and function, and the ability to reliably distinguish between various FOXP3-expressing subpopulations can affect the development of successful therapies. This study demonstrates that hypomethylated CpG sites, present in four regions of the FOXP3 locus, CAMTA1 and FUT7 gene regions, can be used to distinguish several subsets of Treg from conventional CD4+ T lymphocytes (Tcon) in donors of both genders. We describe a previously unreported strand-bias hemimethylation pattern in FOXP3 promoter and TSDR in donors of both genders, with the coding strand being demethylated within promoter and methylated within TSDR in all CD4+ lymphocyte subtypes, whereas the template strand follows the previously described pattern of methylation with both regions being more demethylated in Treg subtypes and mostly methylated in Tcon. This strand-specific approach within the TSDR may prove to be instrumental in correctly defining Treg subsets in health and in disease.
Collapse
Affiliation(s)
- Ekaterina Minskaia
- Faculdade de Medicina da Universidade de Lisboa, Instituto de Medicina Molecular-João Lobo Antunes, Lisbon, Portugal
| | - Barbara C Saraiva
- Faculdade de Medicina da Universidade de Lisboa, Instituto de Medicina Molecular-João Lobo Antunes, Lisbon, Portugal
| | - Maria M V Soares
- Faculdade de Medicina da Universidade de Lisboa, Instituto de Medicina Molecular-João Lobo Antunes, Lisbon, Portugal
| | - Rita I Azevedo
- Faculdade de Medicina da Universidade de Lisboa, Instituto de Medicina Molecular-João Lobo Antunes, Lisbon, Portugal
| | - Ruy M Ribeiro
- Departmento de Biomatemática, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Saumya D Kumar
- Faculdade de Medicina da Universidade de Lisboa, Instituto de Medicina Molecular-João Lobo Antunes, Lisbon, Portugal
| | - Ana I S Vieira
- Faculdade de Medicina da Universidade de Lisboa, Instituto de Medicina Molecular-João Lobo Antunes, Lisbon, Portugal
| | - João F Lacerda
- Faculdade de Medicina da Universidade de Lisboa, Instituto de Medicina Molecular-João Lobo Antunes, Lisbon, Portugal
| |
Collapse
|
126
|
Age-associated methylation change of CHI promoter in herbaceous peony ( Paeonia lactiflora Pall). Biosci Rep 2018; 38:BSR20180482. [PMID: 30061184 PMCID: PMC6137250 DOI: 10.1042/bsr20180482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/22/2018] [Accepted: 06/29/2018] [Indexed: 11/17/2022] Open
Abstract
Chalcone isomerase gene (CHI) is a key gene that regulates the formation of yellow traits in petals. To reveal transcriptional regulatory mechanisms of CHI gene in petals of Paeonia lactiflora, we investigated the CHI expression using qPCR, the pigment content by HPLC, and methylation levels using BSP+Miseq sequencing in ‘Huangjinlun’ variety during different developmental stages including flower-bud stage (S1), initiating bloom (S2), bloom stage (S3), and withering stage (S4). Results showed that the expression level of CHI gene at S2 stage was significantly higher than that at other stages (P<0.05), and at S4 stage was extremely significantly lower than other stages (P<0.01). Besides, total anthocyanin, anthoxanthin, and flavonoid contents in petals presented a similar trend with CHI expression during developmental stages. A total of 16 CpG sites varying methylation levels were detected in CHI gene core promoter region, of which the methylation levels at mC-4 and mC-16 sites were extremely significantly negatively correlated with CHI mRNA expression (P<0.01). mC-16 site is located in the binding region of C/EBPα transcription factor, suggesting that methylation at the mC-16 site may inhibit the binding of C/EBPα to CHI promoter DNA, thereby regulating the tissue-specific expression of CHI gene. Our study revealed the expression pattern of CHI gene in petal tissues of P. lactiflora at different developmental stages, which is related to promoter methylation. Moreover, the important transcription regulation element–C/EBPα was identified, providing theoretical reference for in-depth study on the function of CHI gene in P. lactiflora.
Collapse
|
127
|
Khosravi M, Bidmeshkipour A, Cohen JL, Moravej A, Hojjat-Assari S, Naserian S, Karimi MH. Induction of CD4 +CD25 +FOXP3 + regulatory T cells by mesenchymal stem cells is associated with modulation of ubiquitination factors and TSDR demethylation. Stem Cell Res Ther 2018; 9:273. [PMID: 30359308 PMCID: PMC6203284 DOI: 10.1186/s13287-018-0991-1] [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] [Received: 05/01/2018] [Revised: 08/23/2018] [Accepted: 08/23/2018] [Indexed: 12/11/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are known for their ability to induce the conversion of conventional T cells (Tconvs) into induced regulatory T cells (iTregs) in specific inflammatory contexts. Stable Foxp3 expression plays a major role in the phenotypic and functional stability of iTregs. However, how MSCs induce stable Foxp3 expression remains unknown. Methods We first investigated the role of cell–cell contact and cytokine secretion by bone marrow-derived MSCs (BM-MSCs) on the induction, stability, and suppressive functions of Tregs under various experimental conditions that lead to Foxp3 generation by flow cytometry and ELISA respectively. Second, we studied the effect of MSCs on TRAF6, GRAIL, USP7, STUB1, and UBC13 mRNA expression in CD4+ T cells in correlation with the suppressive function of iTregs by real-time PCR; also, we investigated Foxp3 Treg-specific demethylated region (TSDR) methylation in correlation with Foxp3 stability by the high-resolution melting technique. Third, we studied the effect of ex-vivo-expanded BM-MSCs on the induction of transplant tolerance in a model of fully allogeneic skin transplantation. We further analyzed the cytokine secretion patterns in grafted mice as well as the mRNA expression of ubiquitination genes in CD4+ T cells collected from the spleens of protected mice. Results We found that in-vitro MSC-induced Tregs express high mRNA levels of ubiquitination genes such as TRAF6, GRAIL, and USP7 and low levels of STUB1. Moreover, they have enhanced TSDR demethylation. Infusion of MSCs in a murine model of allogeneic skin transplantation prolonged allograft survival. When CD4+ T cells were harvested from the spleens of grafted mice, we observed that mRNA expression of the Foxp3 gene was elevated. Furthermore, Foxp3 mRNA expression was associated with increased TRAF6, GRAIL, UBC13, and USP7 and decreased STUB1 mRNA levels compared with the levels observed in vitro. Conclusions Our data suggest a possible ubiquitination mechanism by which MSCs convert Tconvs to suppressive and stable iTregs. Electronic supplementary material The online version of this article (10.1186/s13287-018-0991-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maryam Khosravi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.,Inserm, U955, Equipe 21, F-94000, Créteil, France
| | - Ali Bidmeshkipour
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - José L Cohen
- Université Paris-Est, UMR_S955, UPEC, F-94000, Créteil, France.,Inserm, U955, Equipe 21, F-94000, Créteil, France.,UPEC, APHP, Inserm, CIC Biothérapie, Hôpital Henri Mondor, 94010, Créteil, France
| | - Ali Moravej
- Noncommunicable Diseases Research Centre, Fasa University of Medical Sciences, Fasa, Iran
| | - Suzzan Hojjat-Assari
- Institut Français de Recherche et d'Enseignement Supérieur à l'International (IFRES-INT), Paris, France
| | - Sina Naserian
- Université Paris-Est, UMR_S955, UPEC, F-94000, Créteil, France.,Inserm, U955, Equipe 21, F-94000, Créteil, France.,Inserm, U1197, Hôpital Paul Brousse, 94807, Villejuif, France.,SivanCell, Alborz University of Medical Sciences, Alborz, Iran
| | | |
Collapse
|
128
|
Wu Z, Feng H, Cao Y, Huang Y, Dai C, Wu S, Bao W. New Insight into the Molecular Mechanism of the FUT2 Regulating Escherichia coli F18 Resistance in Weaned Piglets. Int J Mol Sci 2018; 19:E3301. [PMID: 30352970 PMCID: PMC6275016 DOI: 10.3390/ijms19113301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 10/10/2018] [Indexed: 11/23/2022] Open
Abstract
Escherichia coli (E. coli) F18 is the main pathogen responsible for post-weaning diarrhea (PWD) in piglets. Resistance to E. coli F18 depends on the expression of the cognate receptors in the intestinal epithelial cells. However, the molecular mechanism of E. coli F18 resistance in weaned piglets remains unclear. Here, we performed a comparative transcriptome study of the duodenal tissue from Sutai E. coli F18 sensitive and resistant pigs by RNA-seq, and pig α(1,2) fucosyltransferase 2 (FUT2) was identified as a host differentially expressed gene controlling the E. coli F18 infection. Function analysis showed that the FUT2 expression was high in the duodenum and jejunum, with higher levels detected in sensitive individuals than in resistant individuals (p < 0.01). Expression levels of FUT2 were upregulated in IPEC-J2 cells after lipopolysaccharide (LPS)-induction or E. coli stimulation. FUT2 knockdown decreased the adhesion of E. coli F18 to IPEC-J2 cells (p < 0.05). FUT2 overexpression markedly increased the adhesion of E. coli F18 to IPEC-J2 cells (p < 0.05 or p < 0.01). Furthermore, the FUT2 mRNA levels correlated with methylation levels of the mC-22 site in the specificity protein 1 (Sp1) transcription factor (p < 0.05). Electrophoretic mobility shift assays (EMSA) showed that Sp1 interacts with the wild-type FUT2 promoter DNA, but not with methylated DNA. Our data suggested that FUT2 methylation at the mC-22 site inhibits Sp1 binding to the FUT2 promoter, thereby reducing FUT2 expression and enhancing E. coli F18 resistance in weaned piglets. These observations highlight FUT2 as a promising new target for combating E. coli F18 susceptibility in weaned piglets.
Collapse
Affiliation(s)
- Zhengchang Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Haiyue Feng
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Yue Cao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Yanjie Huang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Chaohui Dai
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China.
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
129
|
Lee S, Park K, Kim J, Min H, Seong RH. Foxp3 expression in induced regulatory T cells is stabilized by C/EBP in inflammatory environments. EMBO Rep 2018; 19:embr.201845995. [PMID: 30348891 DOI: 10.15252/embr.201845995] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 12/22/2022] Open
Abstract
Proper control of immune responses by Foxp3+ regulatory T cells at inflamed sites is crucial for the prevention of immunopathology. TGF-β-induced Foxp3+ regulatory T (Treg) cells are generated in inflammatory environments as well as in steady-state conditions. Inflammatory cytokines such as IFN-γ and IL-4 have an antagonistic effect on Treg cell conversion. However, it is not known how naive CD4+ T cells overcome the inhibitory environment in inflamed sites to differentiate into Treg cells. Here, we show that CCAAT/enhancer-binding protein (C/EBP) functions as a safeguard that enhances Treg cell generation by dampening the inhibitory effect of IFN-γ and IL-4 on Foxp3 expression. We find that C/EBPβ is induced by retinoic acid and binds to the methyl-CRE sequence in the Foxp3 TSDR to sustain its expression. C/EBPβ-transduced iTreg cells show more potent suppressive activity in mouse disease models. We also reveal that C/EBPβ-transduced human iTreg cells exhibit more enhanced suppressor function. These results establish C/EBP as a new molecular target for enhancing the formation and stability of Treg cells in inflammatory environments.
Collapse
Affiliation(s)
- Sungkyu Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Kyungsoo Park
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Jieun Kim
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Hyungyu Min
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Rho H Seong
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| |
Collapse
|
130
|
Sharabi A, Tsokos MG, Ding Y, Malek TR, Klatzmann D, Tsokos GC. Regulatory T cells in the treatment of disease. Nat Rev Drug Discov 2018; 17:823-844. [DOI: 10.1038/nrd.2018.148] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
131
|
Ferretti AP, Bhargava R, Dahan S, Tsokos MG, Tsokos GC. Calcium/Calmodulin Kinase IV Controls the Function of Both T Cells and Kidney Resident Cells. Front Immunol 2018; 9:2113. [PMID: 30333818 PMCID: PMC6176098 DOI: 10.3389/fimmu.2018.02113] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/28/2018] [Indexed: 12/12/2022] Open
Abstract
Calcium calmodulin kinase IV (CaMK4) regulates multiple processes that significantly contribute to the lupus-related pathology by controlling the production of IL-2 and IL-17 by T cells, the proliferation of mesangial cells, and the function and structure of podocytes. CaMK4 is also upregulated in podocytes from patients with focal segmental glomerulosclerosis (FSGS). In both immune and non-immune podocytopathies, CaMK4 disrupts the structure and function of podocytes. In lupus-prone mice, targeted delivery of a CaMK4 inhibitor to CD4+ T cells suppresses both autoimmunity and the development of nephritis. Targeted delivery though to podocytes averts the deposition of immune complexes without affecting autoimmunity in lupus-prone mice and averts pathology induced by adriamycin in normal mice. Therefore, targeted delivery of a CaMK4 inhibitor to podocytes holds high therapeutic promise for both immune (lupus nephritis) and non-immune (FSGS) podocytopathies.
Collapse
Affiliation(s)
- Andrew P Ferretti
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Shani Dahan
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
132
|
Kumar P, Saini S, Khan S, Surendra Lele S, Prabhakar BS. Restoring self-tolerance in autoimmune diseases by enhancing regulatory T-cells. Cell Immunol 2018; 339:41-49. [PMID: 30482489 DOI: 10.1016/j.cellimm.2018.09.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/14/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022]
Abstract
Self-tolerance, the state of unresponsiveness to self-tissues/antigens, is maintained through central and peripheral tolerance mechanisms, and a breach of these mechanisms leads to autoimmune diseases. Foxp3 + T-regulatory cells (Tregs) play an essential role in suppressing autoimmune response directed against self-antigens and thereby regulate self-tolerance. Natural Tregs are differentiated in the thymus on the basis of their higher TCR-affinity to self-antigens and migrate to the periphery where they maintain peripheral tolerance. In addition, extra-thymic differentiation of induced Tregs can occur in the periphery which can control abrupt immune responses under inflammatory conditions. A defect in Treg cell numbers and/or function is found to be associated with the development of autoimmune disease in several experimental models and human autoimmune diseases. Moreover, augmentation of Tregs has been shown to be beneficial in treating autoimmunity in preclinical models, and Treg based cellular therapy has shown initial promise in clinical trials. However, emerging studies have identified an unstable subpopulation of Tregs which expresses pro-inflammatory cytokines under both homeostatic and autoimmune conditions, as well as in ex vivo cultures. In addition, clinical translation of Treg cellular therapy is impeded by limitations such as lack of easier methods for selective expansion of Tregs and higher cost associated with GMP-facilities required for cell sorting, ex vivo expansion and infusion of ex vivo expanded Tregs. Here, we discuss the recent advances in molecular mechanisms regulating Treg differentiation, Foxp3 expression and lineage stability, the role of Tregs in the prevention of various autoimmune diseases, and critically review their clinical utility for treating human autoimmune diseases.
Collapse
Affiliation(s)
- Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Shikha Saini
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Saad Khan
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Swarali Surendra Lele
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA.
| |
Collapse
|
133
|
Brajic A, Franckaert D, Burton O, Bornschein S, Calvanese AL, Demeyer S, Cools J, Dooley J, Schlenner S, Liston A. The Long Non-coding RNA Flatr Anticipates Foxp3 Expression in Regulatory T Cells. Front Immunol 2018; 9:1989. [PMID: 30319599 PMCID: PMC6167443 DOI: 10.3389/fimmu.2018.01989] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/13/2018] [Indexed: 12/30/2022] Open
Abstract
Mammalian genomes encode a plethora of long non-coding RNA (lncRNA). These transcripts are thought to regulate gene expression, influencing biological processes from development to pathology. Results from the few lncRNA that have been studied in the context of the immune system have highlighted potentially critical functions as network regulators. Here we explored the nature of the lncRNA transcriptome in regulatory T cells (Tregs), a subset of CD4+ T cells required to establish and maintain immunological self-tolerance. The identified Treg lncRNA transcriptome showed distinct differences from that of non-regulatory CD4+ T cells, with evidence of direct shaping of the lncRNA transcriptome by Foxp3, the master transcription factor driving the distinct mRNA profile of Tregs. Treg lncRNA changes were disproportionally reversed in the absence of Foxp3, with an enrichment for colocalisation with Foxp3 DNA binding sites, indicating a direct coordination of transcription by Foxp3 independent of the mRNA coordination function. We further identified a novel lncRNA Flatr, as a member of the core Treg lncRNA transcriptome. Flatr expression anticipates Foxp3 expression during in vitro Treg conversion, and Flatr-deficient mice show a mild delay in in vitro and peripheral Treg induction. These results implicate Flatr as part of the upstream cascade leading to Treg conversion, and may provide clues as to the nature of this process.
Collapse
Affiliation(s)
- Aleksandra Brajic
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Dean Franckaert
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Oliver Burton
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Simon Bornschein
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium.,VIB Cancer Research Center, VIB, Leuven, Belgium
| | - Anna L Calvanese
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | | | - Jan Cools
- VIB Cancer Research Center, VIB, Leuven, Belgium
| | - James Dooley
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Susan Schlenner
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Adrian Liston
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| |
Collapse
|
134
|
Bending D, Ono M. From stability to dynamics: understanding molecular mechanisms of regulatory T cells through Foxp3 transcriptional dynamics. Clin Exp Immunol 2018; 197:14-23. [PMID: 30076771 PMCID: PMC6591142 DOI: 10.1111/cei.13194] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2018] [Indexed: 12/30/2022] Open
Abstract
Studies on regulatory T cells (Treg) have focused on thymic Treg as a stable lineage of immunosuppressive T cells, the differentiation of which is controlled by the transcription factor forkhead box protein 3 (Foxp3). This lineage perspective, however, may constrain hypotheses regarding the role of Foxp3 and Tregin vivo, particularly in clinical settings and immunotherapy development. In this review, we synthesize a new perspective on the role of Foxp3 as a dynamically expressed gene, and thereby revisit the molecular mechanisms for the transcriptional regulation of Foxp3. In particular, we introduce a recent advancement in the study of Foxp3‐mediated T cell regulation through the development of the Timer of cell kinetics and activity (Tocky) system, and show that the investigation of Foxp3 transcriptional dynamics can reveal temporal changes in the differentiation and function of Tregin vivo. We highlight the role of Foxp3 as a gene downstream of T cell receptor (TCR) signalling and show that temporally persistent TCR signals initiate Foxp3 transcription in self‐reactive thymocytes. In addition, we feature the autoregulatory transcriptional circuit for the Foxp3 gene as a mechanism for consolidating Treg differentiation and activating their suppressive functions. Furthermore, we explore the potential mechanisms behind the dynamic regulation of epigenetic modifications and chromatin architecture for Foxp3 transcription. Lastly, we discuss the clinical relevance of temporal changes in the differentiation and activation of Treg.
Collapse
Affiliation(s)
- D Bending
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, UK.,Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - M Ono
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| |
Collapse
|
135
|
Alpinetin exerts anti-colitis efficacy by activating AhR, regulating miR-302/DNMT-1/CREB signals, and therefore promoting Treg differentiation. Cell Death Dis 2018; 9:890. [PMID: 30166541 PMCID: PMC6117360 DOI: 10.1038/s41419-018-0814-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 01/13/2023]
Abstract
Alpinetin, a flavonoid compound extracted from the seeds of Alpinia katsumadai Hayata, has been demonstrated to exert massive biological properties. This study aimed to evaluate the effect of alpinetin on dextran sulfate sodium (DSS)-induced colitis, and elucidate the potential mechanisms. Alpinetin significantly alleviated colitis in mice, accompanied with restored Th17/Treg balance in colons. In vitro, alpinetin directly promoted Treg differentiation but exerted little effect on Th17 differentiation, and the action was in an aryl hydrocarbon receptor (AhR)-dependent manner. It acted as a potential AhR activator, evidenced by increased expression of CYP1A1, dissociation of AhR/HSP90 complexes, AhR nuclear translocation, XRE-driven luciferase reporter gene and DNA-binding activity of AhR/ARNT/XRE in T cells. Furthermore, alpinetin significantly promoted expression of miR-302 but not others, and restrained expression of DNMT-1 and methylation level of Foxp3 promoter region in CD4+ T cells and colons of colitis mice. However, the association of CREB and Foxp3 promoter region but not expression, nuclear translocation and DNA-binding activity of CREB was up-regulated by alpinetin in CD4+ T cells. The relationship of alpinetin-adjusted AhR activation, expressions of miR-302 and DNMT-1, association of CREB and Foxp3 promoter region, and Treg differentiation was confirmed by using CH223191, siAhR, miR-302 inhibitor and pcDNA3.1(+)-mDNMT-1. Finally, CH223191 abolished the amelioration of alpinetin on colitis, induction of Treg cells and regulation of miR-302/DNMT-1/CREB signals in colons of colitis mice. In conclusion, alpinetin ameliorated colitis in mice via activating AhR, regulating miR-302/DNMT-1/CREB signals, therefore promoting Treg differentiation.
Collapse
|
136
|
Belot MP, Castell AL, Le Fur S, Bougnères P. Dynamic demethylation of the IL2RA promoter during in vitro CD4+ T cell activation in association with IL2RA expression. Epigenetics 2018; 13:459-472. [PMID: 30096258 DOI: 10.1080/15592294.2018.1469893] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
IL2RA, a subunit of the high affinity receptor for interleukin-2 (IL2), plays a crucial role in immune homeostasis. Notably, IL2RA expression is induced in CD4+ T cells in response to various stimuli and is constitutive in regulatory T cells (Tregs). We selected for our study 18 CpGs located within cognate regulatory regions of the IL2RA locus and characterized their methylation in naive, regulatory, and memory CD4+ T cells. We found that 5/18 CpGs (notably CpG + 3502) show dynamic, active demethylation during the in vitro activation of naive CD4+ T cells. Demethylation of these CpGs correlates with appearance of IL2RA protein at the cell surface. We found no influence of cis located SNP alleles upon CpG methylation. Treg cells show constitutive demethylation at all studied CpGs. Methylation of 9/18 CpGs, including CpG +3502, decreases with age. Our data thus identify CpG +3502 and a few other CpGs at the IL2RA locus as coordinated epigenetic regulators of IL2RA expression in CD4+ T cells. This may contribute to unravel how the IL2RA locus can be involved in immune physiology and pathology.
Collapse
Affiliation(s)
- Marie-Pierre Belot
- a Institut National de la Santé et de la Recherche Médicale UMR1169 , Paris Sud University, Bicêtre Hospital , Le Kremlin-Bicêtre , France.,b Fondation de l'AP-HP pour la Recherche , Paris , France
| | - Anne-Laure Castell
- c Service de Médecine des Adolescents , Paris Sud University, Bicêtre Hospital , Le Kremlin-Bicêtre , France
| | - Sophie Le Fur
- a Institut National de la Santé et de la Recherche Médicale UMR1169 , Paris Sud University, Bicêtre Hospital , Le Kremlin-Bicêtre , France
| | - Pierre Bougnères
- a Institut National de la Santé et de la Recherche Médicale UMR1169 , Paris Sud University, Bicêtre Hospital , Le Kremlin-Bicêtre , France.,c Service de Médecine des Adolescents , Paris Sud University, Bicêtre Hospital , Le Kremlin-Bicêtre , France
| |
Collapse
|
137
|
Remes Lenicov F, Paletta AL, Gonzalez Prinz M, Varese A, Pavillet CE, Lopez Malizia Á, Sabatté J, Geffner JR, Ceballos A. Prostaglandin E2 Antagonizes TGF-β Actions During the Differentiation of Monocytes Into Dendritic Cells. Front Immunol 2018; 9:1441. [PMID: 29988364 PMCID: PMC6023975 DOI: 10.3389/fimmu.2018.01441] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/11/2018] [Indexed: 12/27/2022] Open
Abstract
Inflammatory dendritic cells (DCs) are a distinct subset of DCs that derive from circulating monocytes infiltrating injured tissues. Monocytes can differentiate into DCs with different functional signatures, depending on the presence of environment stimuli. Among these stimuli, transforming growth factor-beta (TGF-β) and prostaglandin E2 (PGE2) have been shown to modulate the differentiation of monocytes into DCs with different phenotypes and functional profiles. In fact, both mediators lead to contrasting outcomes regarding the production of inflammatory and anti-inflammatory cytokines. Previously, we have shown that human semen, which contains high concentrations of PGE2, promoted the differentiation of DCs into a tolerogenic profile through a mechanism dependent on signaling by E-prostanoid receptors 2 and 4. Notably, this effect was induced despite the huge concentration of TGF-β present in semen, suggesting that PGE2 overrides the influence exerted by TGF-β. No previous studies have analyzed the joint actions induced by PGE2 and TGF-β on the function of monocytes or DCs. Here, we analyzed the phenotype and functional profile of monocyte-derived DCs differentiated in the presence of TGF-β and PGE2. DC differentiation guided by TGF-β alone enhanced the expression of CD1a and abrogated LPS-induced expression of IL-10, while differentiation in the presence of PGE2 impaired CD1a expression, preserved CD14 expression, abrogated IL-12 and IL-23 production, stimulated IL-10 production, and promoted the expansion of FoxP3+ regulatory T cells in a mixed lymphocyte reaction. Interestingly, DCs differentiated in the presence of TGF-β and PGE2 showed a phenotype and functional profile closely resembling those induced by PGE2 alone. Finally, we found that PGE2 inhibited TGF-β signaling through an action exerted by EP2 and EP4 receptors coupled to cyclic AMP increase and protein kinase A activity. These results indicate that PGE2 suppresses the influence exerted by TGF-β during DC differentiation, imprinting a tolerogenic signature. High concentrations of TGF-β and PGE2 are usually found in infectious, autoimmune, and neoplastic diseases. Our observations suggest that in these scenarios PGE2 might play a mandatory role in the acquisition of a regulatory profile by DCs.
Collapse
Affiliation(s)
- Federico Remes Lenicov
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Luz Paletta
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melina Gonzalez Prinz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Augusto Varese
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Clara E Pavillet
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Álvaro Lopez Malizia
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Sabatté
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge Raul Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Ceballos
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
138
|
Lamble AJ, Lind EF. Targeting the Immune Microenvironment in Acute Myeloid Leukemia: A Focus on T Cell Immunity. Front Oncol 2018; 8:213. [PMID: 29951373 PMCID: PMC6008423 DOI: 10.3389/fonc.2018.00213] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022] Open
Abstract
Immunotherapies, such as chimeric antigen receptor T cells, bispecific antibodies, and immune checkpoint inhibitors, have emerged as promising modalities in multiple hematologic malignancies. Despite the excitement surrounding immunotherapy, it is currently not possible to predict which patients will respond. Within solid tumors, the status of the immune microenvironment provides valuable insight regarding potential responses to immune therapies. Much less is known about the immune microenvironment within hematologic malignancies but the characteristics of this environment are likely to serve a similar predictive role. Acute myeloid leukemia (AML) is the most common hematologic malignancy in adults, and only 25% of patients are alive 5 years following their diagnosis. There is evidence that manipulation of the immune microenvironment by leukemia cells may play a role in promoting therapy resistance and disease relapse. In addition, it has long been documented that through modulation of the immune system following allogeneic bone marrow transplant, AML can be cured, even in patients with the highest risk disease. These concepts, along with the poor prognosis associated with this disease, have encouraged many groups to start exploring the utility of novel immune therapies in AML. While the implementation of these therapies into clinical trials for AML has been supported by preclinical rationale, many questions still exist surrounding their efficacy, tolerability, and the overall optimal approach. In this review, we discuss what is known about the immune microenvironment within AML with a specific focus on T cells and checkpoints, along with their implications for immune therapies.
Collapse
Affiliation(s)
- Adam J Lamble
- Pediatric Hematology/Oncology, Seattle Children's Hospital, Seattle, WA, United States
| | - Evan F Lind
- Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
139
|
STAT3-mediated epigenetic silencing of FOXP3 in LADA T cells is regulated through HDAC5 and DNMT1. Clin Immunol 2018; 191:116-125. [DOI: 10.1016/j.clim.2017.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/16/2017] [Accepted: 12/04/2017] [Indexed: 01/09/2023]
|
140
|
Sharma R, Kinsey GR. Regulatory T cells in acute and chronic kidney diseases. Am J Physiol Renal Physiol 2018; 314:F679-F698. [PMID: 28877881 PMCID: PMC6031912 DOI: 10.1152/ajprenal.00236.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/18/2017] [Accepted: 09/04/2017] [Indexed: 02/07/2023] Open
Abstract
Foxp3-expressing CD4+ regulatory T cells (Tregs) make up one subset of the helper T cells (Th) and are one of the major mechanisms of peripheral tolerance. Tregs prevent abnormal activation of the immune system throughout the lifespan, thus protecting from autoimmune and inflammatory diseases. Recent studies have elucidated the role of Tregs beyond autoimmunity. Tregs play important functions in controlling not only innate and adaptive immune cell activation, but also regulate nonimmune cell function during insults and injury. Inflammation contributes to a multitude of acute and chronic diseases affecting the kidneys. This review examines the role of Tregs in pathogenesis of renal inflammatory diseases and explores the approaches for enhancing Tregs for prevention and therapy of renal inflammation.
Collapse
Affiliation(s)
- Rahul Sharma
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia , Charlottesville, Virginia
| | - Gilbert R Kinsey
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia , Charlottesville, Virginia
| |
Collapse
|
141
|
Miao J, Zhu P. Functional Defects of Treg Cells: New Targets in Rheumatic Diseases, Including Ankylosing Spondylitis. Curr Rheumatol Rep 2018; 20:30. [PMID: 29663162 DOI: 10.1007/s11926-018-0729-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW This study aims to review the advances of Treg cell biology, the functional defects of Treg cells, and the potential strategies for the experimental, preclinical or clinical application of Treg cell therapy in the context of autoimmune/immune-mediated rheumatic diseases. RECENT FINDINGS CD4+CD25+ regulatory T (Treg) cells are a phenotypically and functionally heterogeneous subset of lymphocytes that prevent a variety of autoimmune diseases. As in many autoimmune diseases, the functional defects of Treg cells are supposed to play relevant roles in the pathogenesis and development of systemic lupus erythematosus, rheumatoid arthritis, ankylosing spondylitis, and other autoimmune/immune-mediated rheumatic diseases. Consequently, manipulation and modulation of Treg cells represent a potent strategy for therapeutic benefit in many such diseases. A further understanding of the functional defects of Treg cells in rheumatic diseases will contribute to find new targets and therapies in rheumatic diseases, including ankylosing spondylitis.
Collapse
Affiliation(s)
- Jinlin Miao
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an, 710032, Shaanxi Province, People's Republic of China.
| |
Collapse
|
142
|
von Borstel A, Sanders JS, Rutgers A, Stegeman CA, Heeringa P, Abdulahad WH. Cellular immune regulation in the pathogenesis of ANCA-associated vasculitides. Autoimmun Rev 2018; 17:413-421. [DOI: 10.1016/j.autrev.2017.12.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/03/2017] [Indexed: 12/17/2022]
|
143
|
El Fakih R, Komrokji R, Shaheen M, Almohareb F, Rasheed W, Hassanein M. Azacitidine Use for Myeloid Neoplasms. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:e147-e155. [DOI: 10.1016/j.clml.2018.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/23/2018] [Accepted: 02/02/2018] [Indexed: 01/08/2023]
|
144
|
Apert C, Romagnoli P, van Meerwijk JPM. IL-2 and IL-15 dependent thymic development of Foxp3-expressing regulatory T lymphocytes. Protein Cell 2018; 9:322-332. [PMID: 28540653 PMCID: PMC5876181 DOI: 10.1007/s13238-017-0425-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/04/2017] [Indexed: 12/15/2022] Open
Abstract
Immunosuppressive regulatory T lymphocytes (Treg) expressing the transcription factor Foxp3 play a vital role in the maintenance of tolerance of the immune-system to self and innocuous non-self. Most Treg that are critical for the maintenance of tolerance to self, develop as an independent T-cell lineage from common T cell precursors in the thymus. In this organ, their differentiation requires signals from the T cell receptor for antigen, from co-stimulatory molecules, as well as from cytokine-receptors. Here we focus on the cytokines implicated in thymic development of Treg, with a particular emphasis on the roles of interleukin-2 (IL-2) and IL-15. The more recently appreciated involvement of TGF-β in thymic Treg development is also briefly discussed. Finally, we discuss how cytokine-dependence of Treg development allows for temporal, quantitative, and potentially qualitative modulation of this process.
Collapse
Affiliation(s)
- Cécile Apert
- CPTP, Université de Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Paola Romagnoli
- CPTP, Université de Toulouse, CNRS, Inserm, UPS, Toulouse, France.
| | | |
Collapse
|
145
|
Induction of Interferon Kappa in Human Papillomavirus 16 Infection by Transforming Growth Factor Beta-Induced Promoter Demethylation. J Virol 2018; 92:JVI.01714-17. [PMID: 29437968 DOI: 10.1128/jvi.01714-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/30/2018] [Indexed: 12/18/2022] Open
Abstract
Persistent high-risk human papillomavirus (HPV) infection is the major causal factor in cervical and other anogenital cancers. Because there are currently no therapeutics capable of preventing neoplastic progression of HPV infections, understanding the mechanisms of HPV-mediated persistence, including immune evasion, is a major research priority. The multifunctional growth factor transforming growth factor beta (TGFβ) has been shown to inhibit expression of early viral transcripts from cells harboring integrated HPV genomes or cells infected with retroviruses expressing HPV oncoproteins. However, the mechanism of TGFβ-induced inhibition has not been fully defined. In this study, we have observed a previously uncharacterized ability of TGFβ to repress the differentiation-induced upregulation of late HPV16 gene expression. In addition, interferon kappa (IFN-κ), a keratinocyte-specific, constitutively expressed cytokine suppressed by differentiation, can be transcriptionally induced by TGFβ1. TGFβ-mediated IFN-κ transcription only occurs in cells containing HPV16, and this is due to TGFβ1-mediated reversal of HPV-induced methylation of the IFN-κ promoter through active DNA demethylation mediated by thymine DNA glycosylase (TDG). This novel interaction between growth factor and innate immune signaling may shed light on the mechanisms of HPV persistence and how the virus manipulates both immune and growth factor signaling to promote its life cycle.IMPORTANCE Persistent infection by high-risk HPVs is the primary risk factor for development of HPV-induced cancers. Persistence involves viral evasion of the immune response, including the IFN response. HPV is also known to suppress TGFβ signaling, which inhibits viral gene expression. Here, we show that the TGFβ and IFN pathways are interrelated in the context of HPV16 infection through the upregulation of IFN-κ by TGFβ. The ability of TGFβ to induce IFN-κ promoter demethylation and transcriptional activation provides a new explanation for why HPV has evolved mechanisms to inhibit TGFβ in infected cells.
Collapse
|
146
|
İscan B, Tuzun F, Eroglu Filibeli B, Cilekar Micili S, Ergur BU, Duman N, Ozkan H, Kumral A. Effects of maternal folic acid supplementation on airway remodeling and allergic airway disease development. J Matern Fetal Neonatal Med 2018; 32:2970-2978. [PMID: 29587542 DOI: 10.1080/14767058.2018.1452904] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Objective: Maternal folic acid supplementation has been recommended prior to and during the first trimester of pregnancy to reduce the risk of infant neural tube defects. However, an uncertain relationship between folic acid supplementation during pregnancy and development of childhood asthma exists. Recent data show a methyl donor-rich diet could increase the risk of developing allergic airway disease through DNA methylation and aberrant gene transcription. This study evaluated the effect of folic acid supplementation during pregnancy on airway remodeling and allergic airway disease vulnerability in a mouse asthma model. Methods: BALB/c mice were divided into four groups according to gestational folic acid supplementation and postnatal ovalbumin (OVA) exposure: Group 1 (whole pregnancy folic acid supplementation + OVA-exposed group), Group 2 (first gestational week folic acid supplementation + OVA-exposed group), Group 3 (no folic acid supplementation + OVA-exposed group), and Group 4 (control group). Offspring were sacrificed on day 45 for immunohistological and ultrastructural tests. Results: In OVA challenged groups, folic acid supplementation led to a thicker epithelial and subepithelial smooth muscle layer than in the unsupplemented group. Moreover, folic acid supplementation during whole pregnancy (Group 1) was associated with a thicker epithelial and subepithelial smooth muscle layer than folic acid supplementation during the first week of pregnancy (Group 2), suggesting a duration-response relationship. Electron microscopic imaging revealed that structural changes including the loss of epithelial integrity, thickening of basement membrane, and subepithelial fibrosis were more prominent in the folic acid supplementation groups. Conclusions: This study suggested that maternal folic acid supplementation during pregnancy affects airway remodeling and increases the allergic responses induced by ovalbumin challenge in offspring. In addition, the effect size increased as the duration and cumulative dose increased.
Collapse
Affiliation(s)
- Burcin İscan
- a Division of Neonatology, Department of Pediatrics , Dokuz Eylul University School of Medicine, Division of Neonatology , Izmir , Turkey
| | - Funda Tuzun
- a Division of Neonatology, Department of Pediatrics , Dokuz Eylul University School of Medicine, Division of Neonatology , Izmir , Turkey
| | - Berna Eroglu Filibeli
- a Division of Neonatology, Department of Pediatrics , Dokuz Eylul University School of Medicine, Division of Neonatology , Izmir , Turkey
| | - Serap Cilekar Micili
- b Department of Histology , Dokuz Eylul University School of Medicine , Izmir , Turkey
| | - Bekir Ugur Ergur
- b Department of Histology , Dokuz Eylul University School of Medicine , Izmir , Turkey
| | - Nuray Duman
- a Division of Neonatology, Department of Pediatrics , Dokuz Eylul University School of Medicine, Division of Neonatology , Izmir , Turkey
| | - Hasan Ozkan
- a Division of Neonatology, Department of Pediatrics , Dokuz Eylul University School of Medicine, Division of Neonatology , Izmir , Turkey
| | - Abdullah Kumral
- a Division of Neonatology, Department of Pediatrics , Dokuz Eylul University School of Medicine, Division of Neonatology , Izmir , Turkey
| |
Collapse
|
147
|
Russell‐Hallinan A, Watson CJ, Baugh JA. Epigenetics of Aberrant Cardiac Wound Healing. Compr Physiol 2018; 8:451-491. [DOI: 10.1002/cphy.c170029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
148
|
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
|
149
|
Choi YS, Jung MK, Lee J, Choi SJ, Choi SH, Lee HW, Lee JJ, Kim HJ, Ahn SH, Lee DH, Kim W, Park SH, Huh JR, Kim HP, Park JY, Shin EC. Tumor Necrosis Factor-producing T-regulatory Cells Are Associated With Severe Liver Injury in Patients With Acute Hepatitis A. Gastroenterology 2018; 154:1047-1060. [PMID: 29229400 PMCID: PMC5847455 DOI: 10.1053/j.gastro.2017.11.277] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIMS CD4+CD25+Foxp3+ T-regulatory (Treg) cells control immune responses and maintain immune homeostasis. However, under inflammatory conditions, Treg cells produce cytokines that promote inflammation. We investigated production of tumor necrosis factor (TNF) by Treg cells in patients with acute hepatitis A (AHA), and examined the characteristics of these cells and association with clinical factors. METHODS We analyzed blood samples collected from 63 patients with AHA at the time of hospitalization (and some at later time points) and 19 healthy donors in South Korea. Liver tissues were collected from patients with fulminant AHA during liver transplantation. Peripheral blood mononuclear cells were isolated from whole blood and lymphocytes were isolated from liver tissues and analyzed by flow cytometry. Cytokine production from Treg cells (CD4+CD25+Foxp3+) was measured by immunofluorescence levels following stimulation with anti-CD3 and anti-CD28. Epigenetic stability of Treg cells was determined based on DNA methylation patterns. Phenotypes of Treg cells were analyzed by flow cytometry and an RORγt inhibitor, ML-209, was used to inhibit TNF production. Treg cell suppression assay was performed by co-culture of Treg-depleted peripheral blood mononuclear cells s and isolated Treg cells. RESULTS A higher proportion of CD4+CD25+Foxp3+ Treg cells from patients with AHA compared with controls produced TNF upon stimulation with anti-CD3 and anti-CD28 (11.2% vs 2.8%). DNA methylation analysis confirmed the identity of the Treg cells. TNF-producing Treg cells had features of T-helper 17 cells, including up-regulation of RORγt, which was required for TNF production. The Treg cells had reduced suppressive functions compared with Treg cells from controls. The frequency of TNF-producing Treg cells in AHA patients' blood correlated with their serum level of alanine aminotransferase. CONCLUSIONS Treg cells from patients with AHA have altered functions compared with Treg cells from healthy individuals. Treg cells from patients with AHA produce higher levels of TNF, gain features of T-helper 17 cells, and have reduced suppressive activity. The presence of these cells is associated with severe liver injury in patients with AHA.
Collapse
Affiliation(s)
- Yoon Seok Choi
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea,Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Min Kyung Jung
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jeewon Lee
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seong Jin Choi
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Sung Hoon Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Woong Lee
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Jong-Joo Lee
- Department of Environmental Medical Biology, Institute of Tropical Medicine, and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyung Joon Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong Hyeon Lee
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Won Kim
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Su-Hyung Park
- Laboratory of Translational Immunology and Vaccinology, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jun R. Huh
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hyoung-Pyo Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
150
|
Tsai HC, Velichko S, Lee S, Wu R. Cholera toxin enhances interleukin-17A production in both CD4 + and CD8 + cells via a cAMP/protein kinase A-mediated interleukin-17A promoter activation. Immunology 2018; 154:500-509. [PMID: 29377102 DOI: 10.1111/imm.12900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/07/2018] [Accepted: 01/18/2018] [Indexed: 12/26/2022] Open
Abstract
Cholera toxin (CT) is a bacterial component that increases intracellular cAMP levels in host cells and suppresses T-cell activation. Recently, CT was reported to induce T helper type 17-skewing dendritic cells and activate interleukin-17A (IL-17A) production in CD4+ T cells through a cAMP-dependent pathway. However, the underlying mechanism by which cAMP regulates IL-17A production in T cells is not completely defined. In this study, we took advantage of a small molecule protein kinase A (PKA) inhibitor (H89) and different cAMP analogues: a PKA-specific activator (N6-benzoyl-adenosine-cAMP), an exchange protein activated by cAMP-specific activator (Rp-8-chlorophenylthio-2'-O-methyl cAMP), and a PKA inhibitor (Rp-8-bromo-cAMP), to elucidate the signalling cascade of cAMP in IL-17A regulation in T cells. We found that CT induced IL-17A production and IL-17A promoter activity in activated CD4+ T cells through a cAMP/PKA pathway. Moreover, this regulation was via cAMP-response element binding protein (CREB) -mediated transcriptional activation by using the transfection of an IL-17A promoter-luciferase reporter construct and CREB small interfering RNA in Jurkat cells. Also, we showed that CREB bound to the CRE motif located at -183 of the IL-17A promoter in vitro. Most interestingly, not only in CD4+ T cells, CT also enhanced cAMP/PKA-dependent IL-17A production and CREB phosphorylation in CD8+ T cells. In conclusion, our data suggest that CT induces an IL-17A-dominated immune microenvironment through the cAMP/PKA/CREB signalling pathway. Our study also highlights the potentials of CT and cAMP in modulating T helper type 17 responses in vivo.
Collapse
Affiliation(s)
- Hsing-Chuan Tsai
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, CA, USA.,Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Sharlene Velichko
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, CA, USA
| | - Shanshan Lee
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, CA, USA
| | - Reen Wu
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, CA, USA
| |
Collapse
|