151
|
Xie MM, Dent AL. Unexpected Help: Follicular Regulatory T Cells in the Germinal Center. Front Immunol 2018; 9:1536. [PMID: 30013575 PMCID: PMC6036241 DOI: 10.3389/fimmu.2018.01536] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/21/2018] [Indexed: 01/22/2023] Open
Abstract
Follicular helper T (Tfh) cells are necessary for germinal center (GC) formation and within the GC, provide key signals to B cells for their differentiation into plasmablasts and plasma cells that secrete high-affinity and isotype-switched antibody (Ab). A specialized subset of Foxp3+ T cells termed T follicular regulatory (Tfr) cells, also regulate the differentiation of Ab-secreting cells from the GC. Tfr-cell function in the GC is not well understood, however, the dominant paradigm currently is that Tfr cells repress excessive Tfh and GC B cell proliferation and help promote stringent selection of high-affinity B cells. A mouse model where the Bcl6 gene is specifically deleted in Foxp3+ T cells (Bcl6FC mice) allows the study of Tfr cell function with more precision than other approaches. Studies with this model have shown that Tfr cells play a key role in maintaining GC B cell proliferation and Ab levels. Part of the mechanism for this positive "helper" effect of Tfr cells on the GC is Tfr cell-derived IL-10, which can promote B cell growth and entry into the dark zone of the GC. Recent studies on Tfr cells support a new paradigm for Tfr cell function in the GC reaction. Here, we review studies on Tfr cell functions and discuss the evidence that Tfr cells can have a major helper role in the GC-dependent Ab response.
Collapse
Affiliation(s)
- Markus M Xie
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
152
|
Liu Y, Qin T, Zhao X, Dong S, Zhu J, Peng D, Zhong J, Li T, Chen X. Skewed balance of regulatory T cell and inflammatory T cell in IL-17 defect with human metapneumovirus infection. Cell Immunol 2018; 331:161-167. [PMID: 30077332 DOI: 10.1016/j.cellimm.2018.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/25/2018] [Accepted: 06/18/2018] [Indexed: 01/01/2023]
Abstract
Human metapneumovirus (hMPV) is a common cause of respiratory infections in children. However, the precise mechanisms underlying the development of hMPV-induced pulmonary pathology remain unknown. Studies show that IL-17 plays an important role in some inflammatory diseases of the airways, including asthma and chronic obstructive pulmonary disease. Here, we generated an IL-17 KO murine model of hMPV infection and used it to characterize the role of IL-17 hMPV-induced pulmonary inflammation. The results demonstrated that the defect in IL-17 resulted in less neutrophil influx into the lungs, along with reduced ventilatory function. Meanwhile, viral infection in IL-17 KO mice increased regulatory T cells (Tregs) and reduced Th1 and Th2 cells in the lung, suggesting that lack of IL-17 skews the immune response in the lung toward an anti-inflammatory profile, as exhibited by a greater number of Treg cells and fewer Th1 and Th2 cells.
Collapse
Affiliation(s)
- Yuhang Liu
- Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Tao Qin
- Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiaodong Zhao
- Research Center for Immunologic and Infectious Diseases, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Division of Immunology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing 400014, China
| | - Shifang Dong
- Division of Flow Cytometry, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jin Zhu
- Division of Pathology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Donghong Peng
- Division of Respiratory, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jiarong Zhong
- General Medical Wards, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Tingyu Li
- Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xin Chen
- General Medical Wards, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
153
|
Abstract
Although mutations of the dystrophin gene are the causative defect in Duchenne muscular dystrophy (DMD) patients, secondary disease processes such as inflammation contribute greatly to the pathogenesis of DMD. Genetic and histological studies have shown that distinct facets of the immune system promote muscle degeneration or regeneration during muscular dystrophy through mechanisms that are only beginning to be defined. Although histological methods have allowed the enumeration and localization of immune cells within dystrophic muscle, they are limited in their ability to assess the full spectrum of phenotypic states of an immune cell population and its functional characteristics. This chapter highlights flow cytometry methods for the isolation and functional study of immune cell populations from muscle of the mdx mouse model of DMD. We include a detailed description of preparing single-cell suspensions of dystrophic muscle that maintain the integrity of cell-surface markers used to identify macrophages, eosinophils, group 2 innate lymphoid cells, and regulatory T cells. This method complements the battery of histological assays that are currently used to study the role of inflammation in muscular dystrophy, and provides a platform capable of being integrated with multiple downstream methodologies for the mechanistic study of immunity in muscle degenerative diseases.
Collapse
|
154
|
Wang CY, Ding HZ, Tang X, Li ZG. Effect of Liuweibuqi capsules on CD4 +CD25 +Foxp3 + regulatory T cells, helper T cells and lung function in patients with stable chronic obstructive pulmonary disease complicated with lung Qi deficiency. J Thorac Dis 2018; 10:2700-2711. [PMID: 29997932 DOI: 10.21037/jtd.2018.04.110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Chronic obstructive pulmonary disease (COPD) is predicted to become the fifth leading cause of disability and the third leading cause of death around the world by 2020. Though it is potentially treatable and preventable, evidence of brain structural alterations in COPD remains sparse and conflicting. We aim to investigate the effect of Liuweibuqi capsules on CD4+CD25+ Forkhead box protein 3+ (Foxp3+) regulatory T cells (Tregs), helper T cells (Th) and lung function in patients with stable COPD complicated with lung Qi deficiency. Methods COPD patients with lung Qi deficiency [458] were assigned into non-smoking COPD (NS-COPD), non-smoking control (NS-control), smoking COPD (S-COPD) and smoking control (S-control) groups, and healthy volunteers [245] into the non-smoking healthy (NSH) and smoking healthy (SH) groups. Levels of inflammatory cytokines were detected by Enzyme-linked immunoassay (ELISA). Contents of inflammatory cells, inflammatory marker, and CD4+CD25+Fox3+Tregs were measured by flow cytometry. FEV1/FVC (%) and FEV1 (%) were detected by pulmonary function test apparatus. Correlation between FEV1 (%) and Th1, Th2, Th17, Th1/Th2 or CD4+CD25+Fox3+Tregs was analyzed by Spearman rank correlation test. The related factors affecting treatment efficacy was assessed by logistic analysis. Results COPD patients and smoking people showed higher level of INF-γ, IL-4, IL-17, Th1, Th2, Th17 and Th1/Th2 but lower level of CD4+CD25+Fox3+Tregs. Liuweibuqi capsules could decrease level of inflammatory cells, cytokines, and markers (especially Th17 and IL-17), and increase level of CD4+CD25+Fox3+Tregs. FEV1 (%) negatively correlated with Th1, Th2, Th17 and Th1/Th2 but positively correlated with CD4+CD25+Fox3+Tregs, and smoking may strengthen their correlation, but Liuweibuqi capsules may weaker their correlation. Levels of inflammatory cytokines, cells, marker, CD4+CD25+Fox3+Tregs, FEV1/FVC (%), FEV1 (%), smoking and Liuweibuqi capsules are factors affecting efficacy. Conclusions Taken together, our data support the notion that smoking is an important factor to induce and aggravate COPD. Liuweibuqi capsules could stimulate proliferation of CD4+CD25+Fox3+Tregs and decrease Th17 expression to improve the lung function in stable COPD patients with lung Qi deficiency, and it had obvious efficacy for smoking COPD patients.
Collapse
Affiliation(s)
- Cheng-Yang Wang
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Huan-Zhang Ding
- Graduate School of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xiao Tang
- Graduate School of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ze-Geng Li
- Anhui Academy, Chinese Medicine, Hefei 230012, China
| |
Collapse
|
155
|
Revealing the specificity of regulatory T cells in murine autoimmune diabetes. Proc Natl Acad Sci U S A 2018; 115:5265-5270. [PMID: 29712852 DOI: 10.1073/pnas.1715590115] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Regulatory T cells (Tregs) control organ-specific autoimmunity in a tissue antigen-specific manner, yet little is known about their specificity in a natural repertoire. In this study, we used the nonobese diabetic (NOD) mouse model of autoimmune diabetes to investigate the antigen specificity of Tregs present in the inflamed tissue, the islets of Langerhans. Compared with Tregs present in spleen and lymph node, Tregs in the islets showed evidence of antigen stimulation that correlated with higher proliferation and expression of activation markers CD103, ICOS, and TIGIT. T cell receptor (TCR) repertoire profiling demonstrated that islet Treg clonotypes are expanded in the islets, suggesting localized antigen-driven expansion in inflamed islets. To determine their specificity, we captured TCRαβ pairs from islet Tregs using single-cell TCR sequencing and found direct evidence that some of these TCRs were specific for islet-derived antigens including insulin B:9-23 and proinsulin. Consistently, insulin B:9-23 tetramers readily detected insulin-specific Tregs in the islets of NOD mice. Lastly, islet Tregs from prediabetic NOD mice were effective at preventing diabetes in Treg-deficient NOD.CD28-/- recipients. These results provide a glimpse into the specificities of Tregs in a natural repertoire that are crucial for opposing the progression of autoimmune diabetes.
Collapse
|
156
|
Guo H, Xun L, Zhang R, Gou X. Ratio of CD147 high/CD147 low in CD4 +CD25 + T cells: A potential biomarker for early diagnosis and prediction of response to therapy for autoimmune diseases. Med Hypotheses 2018; 115:1-4. [PMID: 29685186 DOI: 10.1016/j.mehy.2018.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/19/2018] [Indexed: 12/16/2022]
Abstract
Regulatory T cell (Treg cell) is an important immunosuppressive T cell subset and plays a dominant role in maintaining the immune balance in vivo. The function defects in Treg cells have been involved in the pathogenesis of many autoimmune diseases. The detection of Treg cell suppressive function is important for early diagnosis and prediction of response to treatment for autoimmune diseases. The traditional detection of Treg cell suppressive function needs at least 20 mL peripheral blood sample of patients and the results would be got in sixth day, therefore, it could not be widely applied in clinical. However, to find fast and simple detection method is very important. CD147 is a transmembrane protein and its expression is related to Treg cell suppressive function. Recent research has shown that the Treg cells with high CD147 expression have stronger suppressive function than which with low CD147 expression. In this work, we detected the ratio of CD147high/CD147low in CD4+CD25+ T cells in patients with active AS using fluorescence-activated cell sorter (FACS). The results show the ratio of CD147high/CD147low decreased obviously in patients with active AS compared with healthy controls, which reflects the suppressive function deficit of Treg cell. In the same time, the detection of the ratio of CD147high/CD147low needs only 150 μL peripheral blood sample and the result would be got in 4 h. We therefore hypothesize that the ratio of CD147high/CD147low is a good indicator for the Treg cell function, and it is especially suitable for early diagnosis and prediction of response to therapy targeted recovering Treg cell function in autoimmune diseases.
Collapse
Affiliation(s)
- Huifang Guo
- School of Basic Medical Science & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an 710021, China
| | - Liru Xun
- Shaanxi Provincial People's Hospital Affiliated to Xi'an Medical University, Xi'an 710068, China
| | - Ruisan Zhang
- School of Basic Medical Science & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an 710021, China
| | - Xingchun Gou
- School of Basic Medical Science & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an 710021, China.
| |
Collapse
|
157
|
Ratay ML, Balmert SC, Bassin EJ, Little SR. Controlled release of an HDAC inhibitor for reduction of inflammation in dry eye disease. Acta Biomater 2018. [PMID: 29526828 DOI: 10.1016/j.actbio.2018.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dry eye disease (DED), also known as keratoconjunctivitis sicca, is an ocular surface disease characterized by T-cell-mediated inflammation. Current therapeutics, such as immunosuppressive agents, act to suppress the clinical signs and inflammation. However, long-term usage of these treatments can cause severe side effects. In this study, we present an alternative therapeutic approach that utilizes a histone deacetylase inhibitor (HDACi) to regulate transcription of a variety of immunomodulatory genes. Specifically, HDACi have emerged as a potential anti-inflammatory agent, which can modulate the functions of a subset of suppressive T lymphocytes known as regulatory T cells (Tregs), enhancing FoxP3 acetylation and subsequently guarding the transcription factor from proteasomal degradation. Here, a specific HDACi known as SAHA (suberoylanilide hydroxamic acid) was formulated to controllably release in the lacrimal gland. Intralacrimal gland injection of PLGA-based SAHA microspheres prevented clinical signs of DED in mice with Concanavalin A-induced DED, reduced expression of pro-inflammatory cytokines, and increased expression of FoxP3 in the lacrimal glands. Murine T cell culture experiments also revealed that SAHA decreased effector T cell proliferation and enhanced suppressive function of Tregs in co-cultures of Tregs and effector T cells. STATEMENT OF SIGNIFICANCE In this study, we demonstrate a therapeutic approach that utilizes a histone deactylase inhibitor (HDACi) to regulate transcription of a variety of immunomodulatory genes. HDACi have emerged as a potential anti-inflammatory agent, which can modulate the functions of a subset of suppressive T lymphocytes known as regulatory T cells (Tregs). Here, HDACi microspheres composed of a biocompatible and biodegradable polymer (poly(lactic-co-glycolic acid) (PLGA)), were able to locally release the HDACi and prevent clinical signs of DED. This work is timely given the recent shift in treatments of DED towards immunological based therapies to reduce ocular inflammation. However, notably, many of these treatments require large amounts of drug, and non-specifically suppress the immune system, leading to several systemic side effects. Instead of merely suppressing or blocking inflammation, the formulation described herein intends to balance the microenvironment promoting immunological homeostasis. This particular drug delivery system may also have broad implications in the field of inflammatory mediated ocular disorders such as uveitis, Sjögren's syndrome, allergic conjunctivitis.
Collapse
Affiliation(s)
- Michelle L Ratay
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Stephen C Balmert
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Ethan J Bassin
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Steven R Little
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15216, United States; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
158
|
Affiliation(s)
- Krung Phiwpan
- School of Allied Health Sciences, University of Phayao, Phayao province, Thailand
| | - Xuyu Zhou
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
159
|
Crossfield L, Fortuna L, Carling R, Scurrell E, Matas Riera M, Chang YM, Dawson C, Pittaway R, Yeung IY, Thaung C, Priestnall SL, Garden OA. Immunohistochemical characterization of feline lymphoplasmacytic anterior uveitis. Vet Ophthalmol 2018. [PMID: 29517855 DOI: 10.1111/vop.12569] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To characterize the immune cells present in different forms of feline anterior uveitis. SAMPLES Eyes were obtained from 49 cats diagnosed with chronic idiopathic lymphoplasmacytic anterior uveitis, 7 cats with feline infectious peritonitis (FIP), and 9 cats euthanized for nonocular disease. METHODS H&E sections were scored on the level of infiltrate in the anterior uvea. Immunohistochemistry was performed for FoxP3, CD3, and IL-17A, and positive cells were quantified in multiple images of each sample. A generalized estimating equation tested for an association between the level of inflammation and the prevalence of these cell types. RESULTS Cells stained positive for IL-17A in idiopathic uveitis but not in FIP samples. We found significantly fewer FoxP3+ and CD3+ cells in low-grade compared with high-grade inflammation in idiopathic uveitis or FIP samples (P values all <.005), but no difference between FIP and high-grade samples. CONCLUSIONS Idiopathic, but not FIP-associated, uveitis appears to have Th17 cell involvement. The numbers of FoxP3+ and CD3+ T-cells present appear directly correlated; thus, the severity of disease does not appear directly determined by the numbers of regulatory cells.
Collapse
Affiliation(s)
- Lawrence Crossfield
- Department of Clinical Science and Services, Royal Veterinary College, London, UK
| | - Luca Fortuna
- Department of Clinical Science and Services, Royal Veterinary College, London, UK
| | - Rebekah Carling
- Department of Clinical Science and Services, Royal Veterinary College, London, UK
| | | | - Marian Matas Riera
- Department of Clinical Science and Services, Royal Veterinary College, London, UK
| | - Yu-Mei Chang
- Royal Veterinary College Research Support Office, London, UK
| | - Charlotte Dawson
- Department of Clinical Science and Services, Royal Veterinary College, London, UK
| | - Rachael Pittaway
- Department of Pathobiology & Population Sciences, Royal Veterinary College, London, UK
| | - Ian Yl Yeung
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | | | - Simon L Priestnall
- Department of Pathobiology & Population Sciences, Royal Veterinary College, London, UK
| | - Oliver A Garden
- Department of Clinical Science and Services, Royal Veterinary College, London, UK
| |
Collapse
|
160
|
Xie C, Li X, Zhou X, Li Z, Zhang Y, Zhao L, Hao Y, Zhang GX, Guan Y. TGFβ1 transduction enhances immunomodulatory capacity of neural stem cells in experimental autoimmune encephalomyelitis. Brain Behav Immun 2018; 69:283-295. [PMID: 29203425 DOI: 10.1016/j.bbi.2017.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 11/07/2017] [Accepted: 11/30/2017] [Indexed: 10/18/2022] Open
Abstract
Bone marrow-derived neural stem cells (BM-NSCs) have therapeutic effect on EAE, an animal model of multiple sclerosis. However, the beneficial effect is suboptimal due to the limited immunomodulatory capacity of these cells. In this study, we engineered BM-NSCs with inducible TGFβ1, a potent immunosuppressive cytokine, to enhance their anti-inflammatory capacity. We found that i.v. injected TGFβ1-BM-NSCs more effectively suppressed clinical severity, inflammation and demyelination of the central nervous system of EAE mice. Transduction of TGFβ1 resulted in a higher percentage of Tregs and lower percentage of Th1 and Th17 cells in the periphery, with increased production of IL-10, and reduced production of IFN-γ, IL-17 and GM-CSF. Moreover, myelin-specific splenic proliferation was also inhibited more profoundly by TGFβ1-BM-NSCs. We also found that TGFβ1-BM-NSCs have the capacity to switch microglia from M1 to M2 phenotype. On the other hand, transduction of TGFβ1 did not affect proliferative ability and differentiating potential of BM-NSCs in vitro and in vivo. Together, these findings demonstrate that transduction of TGFβ1 significantly enhanced the immunomodulatory capacity of BM-NSCs for EAE treatment, through inducing Tregs and an M2 phenotype of macrophages/microglia, while retaining their capacity for neural cell differentiation. Thus, our study provides an easily accessible, inducible and effective therapy for CNS inflammatory demyelination.
Collapse
Affiliation(s)
- Chong Xie
- Department of Neurology, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, China
| | - Xing Li
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710062 Xi'an, China
| | - Xiajun Zhou
- Department of Neurology, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, China
| | - Zezhi Li
- Department of Neurology, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, China
| | - Yuan Zhang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710062 Xi'an, China
| | - Li Zhao
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710062 Xi'an, China
| | - Yong Hao
- Department of Neurology, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, China
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, PA19107 Philadelphia, USA.
| | - Yangtai Guan
- Department of Neurology, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, China.
| |
Collapse
|
161
|
Grinberg-Bleyer Y, Caron R, Seeley JJ, De Silva NS, Schindler CW, Hayden MS, Klein U, Ghosh S. The Alternative NF-κB Pathway in Regulatory T Cell Homeostasis and Suppressive Function. THE JOURNAL OF IMMUNOLOGY 2018; 200:2362-2371. [PMID: 29459403 DOI: 10.4049/jimmunol.1800042] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022]
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) are essential regulators of immune responses. Perturbation of Treg homeostasis or function can lead to uncontrolled inflammation and autoimmunity. Therefore, understanding the molecular mechanisms involved in Treg biology remains an active area of investigation. It has been shown previously that the NF-κB family of transcription factors, in particular, the canonical pathway subunits, c-Rel and p65, are crucial for the development, maintenance, and function of Tregs. However, the role of the alternative NF-κB pathway components, p100 and RelB, in Treg biology remains unclear. In this article, we show that conditional deletion of the p100 gene, nfkb2, in Tregs, resulted in massive inflammation because of impaired suppressive function of nfkb2-deficient Tregs. Surprisingly, mice lacking RelB in Tregs did not exhibit the same phenotype. Instead, deletion of both relb and nfkb2 rescued the inflammatory phenotype, demonstrating an essential role for p100 as an inhibitor of RelB in Tregs. Our data therefore illustrate a new role for the alternative NF-κB signaling pathway in Tregs that has implications for the understanding of molecular pathways driving tolerance and immunity.
Collapse
Affiliation(s)
- Yenkel Grinberg-Bleyer
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Rachel Caron
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - John J Seeley
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Nilushi S De Silva
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Christian W Schindler
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Matthew S Hayden
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Ulf Klein
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032;
| |
Collapse
|
162
|
Duggleby R, Danby RD, Madrigal JA, Saudemont A. Clinical Grade Regulatory CD4 + T Cells (Tregs): Moving Toward Cellular-Based Immunomodulatory Therapies. Front Immunol 2018; 9:252. [PMID: 29487602 PMCID: PMC5816789 DOI: 10.3389/fimmu.2018.00252] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/29/2018] [Indexed: 12/26/2022] Open
Abstract
Regulatory T cells (Tregs) are CD4+ T cells that are key players of immune tolerance. They are powerful suppressor cells, able to impact the function of numerous immune cells, including key effectors of inflammation such as effector T cells. For this reason, Tregs are an ideal candidate for the development of cell therapy approaches to modulate immune responses. Treg therapy has shown promising results so far, providing key knowledge on the conditions in which these cells can provide protection and demonstrating that they could be an alternative to current pharmacological immunosuppressive therapies. However, a more comprehensive understanding of their characteristics, isolation, activation, and expansion is needed to be able design cost effective therapies. Here, we review the practicalities of making Tregs a viable cell therapy, in particular, discussing the challenges faced in isolating and manufacturing Tregs and defining what are the most appropriate applications for this new therapy.
Collapse
Affiliation(s)
- Richard Duggleby
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| | - Robert David Danby
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom.,Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| | - Aurore Saudemont
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| |
Collapse
|
163
|
Interleukin-37 Ameliorates Coxsackievirus B3-induced Viral Myocarditis by Modulating the Th17/Regulatory T cell Immune Response. J Cardiovasc Pharmacol 2018; 69:305-313. [PMID: 28207428 DOI: 10.1097/fjc.0000000000000476] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Myocarditis is a heterogeneous group of disorders defined by inflammation of the heart muscle with an excessively activated immune response. Numerous interventions have been investigated for the treatment of myocarditis while success is limited. Interleukin-37 (IL-37), a novel member of the IL-1 cytokine family, is a natural inhibitor of innate immunity associated with autoimmune diseases. However, the modulatory effect of IL-37 in myocarditis is unknown. In this study, we investigated the immunological regulation of IL-37 in the coxsackievirus B3-induced model of murine viral myocarditis. The results show that IL-37 significantly ameliorates the signs of myocarditis with increased survival rate and bodyweight, improved histological changes, reduced activities of MB isoenzyme of creatine kinase and cardiac troponin I, and a suppressed response of Th17 cells and enhanced response of regulatory T cells (Tregs) in the spleen. Moreover, IL-37 down-regulates the expression of Th17-related cytokines IL-6 and IL-17A, while promoting Treg-related cytokine IL-10 levels in the heart. Therefore, IL-37 may exhibit anti-inflammatory activity in the murine model of myocarditis by regulating the balance between Th17 and Treg cells, thereby providing a possible novel therapeutic target in myocarditis.
Collapse
|
164
|
Deng Y, Wang F, Hughes T, Yu J. FOXOs in cancer immunity: Knowns and unknowns. Semin Cancer Biol 2018; 50:53-64. [PMID: 29309928 DOI: 10.1016/j.semcancer.2018.01.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 12/26/2017] [Accepted: 01/04/2018] [Indexed: 12/20/2022]
Abstract
In the tumor microenvironment (TME), cancer cells, stromal cells, and immune cells, along with their extracellular factors, have profound effects on either promoting or repressing anti-cancer immunity. Accumulating evidence has shown the paradoxical intrinsic role of the Forkhead box O (FOXO) family of transcription factors in cancer, which can act as a tumor repressor while also maintaining cancer stem cells. FOXOs also regulate cancer immunity. FOXOs promote antitumor activity through negatively regulating the expression of immunosuppressive proteins, such as programmed death 1 ligand 1 (PD-L1), and vascular endothelial growth factor (VEGF) in tumor cells or stromal cells, which can shape an immunotolerant state in the TME. FOXOs also intrinsically control the anti-tumor immune response as well as the homeostasis and development of immune cells, including T cells, B cells, natural killer (NK) cells, macrophages, and dendritic cells. As a cancer repressor, reviving the activity of Foxo1 forces tumor-infiltrating activated regulatory T (Treg) cells to egress from tumor tissues. As a promoter of cancer development, Foxo3 and Foxo1 negatively regulate cytotoxicity of both CD8+ T cells and NK cells against tumor cells. In this review, we focus on the complex role of FOXOs in regulating cancer immunity due to the various roles that they play in cancer cells, stromal cells, and immune cells. We also speculate on some possible additional roles of FOXOs in cancer immunity based on findings regarding FOXOs in non-cancer settings, such as infectious disease.
Collapse
Affiliation(s)
- Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), China.
| | - Fangjie Wang
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), China
| | - Tiffany Hughes
- Comprehensive Cancer Center, The Ohio State University, United States
| | - Jianhua Yu
- Comprehensive Cancer Center, The Ohio State University, United States; Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, United States; The James Cancer Hospital and Solove Research Institute, The Ohio State University, United States.
| |
Collapse
|
165
|
Marshall GP, Cserny J, Perry DJ, Yeh WI, Seay HR, Elsayed AG, Posgai AL, Brusko TM. Clinical Applications of Regulatory T cells in Adoptive Cell Therapies. CELL & GENE THERAPY INSIGHTS 2018; 4:405-429. [PMID: 34984106 PMCID: PMC8722436 DOI: 10.18609/cgti.2018.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Interest in adoptive T-cell therapies has been ignited by the recent clinical success of genetically-modified T cells in the cancer immunotherapy space. In addition to immune targeting for malignancies, this approach is now being explored for the establishment of immune tolerance with regulatory T cells (Tregs). Herein, we will summarize the basic science and clinical results emanating from trials directed at inducing durable immune regulation through administration of Tregs. We will discuss some of the current challenges facing the field in terms of maximizing cell purity, stability and expansion capacity, while also achieving feasibility and GMP production. Indeed, recent advances in methodologies for Treg isolation, expansion, and optimal source materials represent important strides toward these considerations. Finally, we will review the emerging genetic and biomaterial-based approaches on the horizon for directing Treg specificity to augment tissue-targeting and regenerative medicine.
Collapse
Affiliation(s)
| | - Judit Cserny
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Daniel J Perry
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Wen-I Yeh
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Howard R Seay
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Ahmed G Elsayed
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA.,Department of Microbiology and Immunology, Faculty of Medicine, Mansoura University, Egypt
| | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Todd M Brusko
- OneVax LLC, Sid Martin Biotechnology Institute, Alachua, Florida, USA.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| |
Collapse
|
166
|
Pedros C, Canonigo-Balancio AJ, Kong KF, Altman A. Requirement of Treg-intrinsic CTLA4/PKCη signaling pathway for suppressing tumor immunity. JCI Insight 2017; 2:95692. [PMID: 29212947 DOI: 10.1172/jci.insight.95692] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022] Open
Abstract
The ability of Tregs to control the development of immune responses is essential for maintaining immune system homeostasis. However, Tregs also inhibit the development of efficient antitumor responses. Here, we explored the characteristics and mechanistic basis of the Treg-intrinsic CTLA4/PKCη signaling pathway that we recently found to be required for contact-dependent Treg-mediated suppression. We show that PKCη is required for the Treg-mediated suppression of tumor immunity in vivo. The presence of PKCη-deficient (Prkch-/-) Tregs in the tumor microenvironment was associated with a significantly increased expression of the costimulatory molecule CD86 on intratumoral CD103+ DCs, enhanced priming of antigen-specific CD8+ T cells, and greater levels of effector cytokines produced by these cells. Similar to mouse Tregs, the GIT/PAK/PIX complex also operated downstream of CTLA4 and PKCη in human Tregs, and GIT2 knockdown in Tregs promoted antitumor immunity. Collectively, our data suggest that targeting the CTLA4/PKCη/GIT/PAK/PIX signaling pathway in Tregs could represent a novel immunotherapeutic strategy to alleviate the negative impact of Tregs on antitumor immune responses.
Collapse
Affiliation(s)
- Christophe Pedros
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Ann J Canonigo-Balancio
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA.,Pfizer Oncology Research & Development, La Jolla, California, USA
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| |
Collapse
|
167
|
Ratay ML, Bellotti E, Gottardi R, Little SR. Modern Therapeutic Approaches for Noninfectious Ocular Diseases Involving Inflammation. Adv Healthc Mater 2017; 6:10.1002/adhm.201700733. [PMID: 29034584 PMCID: PMC5915344 DOI: 10.1002/adhm.201700733] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/25/2017] [Indexed: 12/12/2022]
Abstract
Dry eye disease, age-related macular degeneration, and uveitis are ocular diseases that significantly affect the quality of life of millions of people each year. In these diseases, the action of chemokines, proinflammatory cytokines, and immune cells drives a local inflammatory response that results in ocular tissue damage. Multiple therapeutic strategies are developed to either address the symptoms or abate the underlying cause of these diseases. Herein, the challenges to deliver drugs to the relevant location in the eye for each of these diseases are reviewed along with current and innovative therapeutic approaches that attempt to restore homeostasis within the ocular microenvironment.
Collapse
Affiliation(s)
- Michelle L. Ratay
- Department of Bioengineering, University of Pittsburgh, 427 Benedum Hall 3700 O’Hara Street Pittsburgh, Pa 15261
| | - Elena Bellotti
- Department of Chemical Engineering, University of Pittsburgh, 427 Benedum Hall 3700 O’Hara Street Pittsburgh, Pa 15261
| | - Riccardo Gottardi
- Department of Chemical Engineering, Department of Orthopedic Surgery, Ri.MED Foundation, 427 Benedum Hall 3700 O’Hara Street Pittsburgh, Pa 15261
| | - Steven R. Little
- Department of Chemical Engineering, Department of Bioengineering, Department of Ophthalmology, Department of Immunology, Department of Pharmaceutical Sciences, The McGowan Institute for Regenerative Medicine, 940 Benedum Hall 3700 O’Hara Street Pittsburgh Pa 15261
| |
Collapse
|
168
|
Update on the Protective Role of Regulatory T Cells in Myocardial Infarction: A Promising Therapy to Repair the Heart. J Cardiovasc Pharmacol 2017; 68:401-413. [PMID: 27941502 DOI: 10.1097/fjc.0000000000000436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial infarction (MI) remains one of the leading causes of heart failure development and death worldwide. To date, interventional and pharmacological therapies are effective in reducing the onset of heart failure and promoting survival. However, progressive maladaptive remodeling post-MI persists in a large fraction of patients resulting in poor prognosis. Immune cell responses and an inflammatory environment largely contribute to adverse cardiac remodeling post-MI. CD4FOXP3 regulatory T cells (Tregs) are known for their immunosuppressive capacity and have been successfully implemented in multiple preclinical studies of permanent and ischemia-reperfusion MI. In this review, we highlight the important cardioprotective role of Tregs at the cardiac tissue, cellular, and molecular level, as well as the most prominent pharmacological venues that could be used to exploit Tregs as a novel therapeutic intervention to lessen myocardial injury post-MI.
Collapse
|
169
|
Tabares P, Berr S, Langenhorst D, Sawitzki B, ten Berge I, Tony HP, Hünig T. Short-term cytokine stimulation reveals regulatory T cells with down-regulated Foxp3 expression in human peripheral blood. Eur J Immunol 2017; 48:366-379. [DOI: 10.1002/eji.201747244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Accepted: 10/05/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Paula Tabares
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| | - Susanne Berr
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| | - Daniela Langenhorst
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| | - Birgit Sawitzki
- Institute of Medical Immunology; Charité University Medicine; Berlin Germany
| | - Ineke ten Berge
- Department of Internal Medicine; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Hans-Peter Tony
- Department of Internal Medicine II; University Hospital of Würzburg; Würzburg Germany
| | - Thomas Hünig
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| |
Collapse
|
170
|
Mauvais-Jarvis F. Are estrogens promoting immune modulation and islet protection in type 1 diabetes? J Diabetes Complications 2017; 31:1563-1564. [PMID: 28890307 DOI: 10.1016/j.jdiacomp.2017.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Franck Mauvais-Jarvis
- Section of Endocrinology & Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
171
|
Aas IB, Austbø L, Falk K, Hordvik I, Koppang EO. The interbranchial lymphoid tissue likely contributes to immune tolerance and defense in the gills of Atlantic salmon. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 76:247-254. [PMID: 28655579 DOI: 10.1016/j.dci.2017.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 06/07/2023]
Abstract
Central and peripheral immune tolerance is together with defense mechanisms a hallmark of all lymphoid tissues. In fish, such tolerance is especially important in the gills, where the intimate contact between gill tissue and the aqueous environment would otherwise lead to continual immune stimulation by innocuous antigens. In this paper, we focus on the expression of genes associated with immune regulation by the interbranchial lymphoid tissue (ILT) in an attempt to understand its role in maintaining immune homeostasis. Both healthy and virus-challenged fish were investigated, and transcript levels were examined from laser-dissected ILT, gills, head kidney and intestine. Lack of Aire expression in the ILT excluded its involvement in central tolerance and any possibility of its being an analogue to the thymus. On the other hand, the ILT appears to participate in peripheral immune tolerance due to its relatively high expression of forkhead box protein 3 (Foxp3) and other genes associated with regulatory T cells (Tregs) and immune suppression.
Collapse
Affiliation(s)
- Ida Bergva Aas
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Lars Austbø
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Knut Falk
- Norwegian Veterinary Institute, 0454 Oslo, Norway
| | - Ivar Hordvik
- Department of Biology, High Technology Centre, University of Bergen, 5006 Bergen, Norway
| | - Erling Olaf Koppang
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway.
| |
Collapse
|
172
|
Chandran S, Tang Q, Sarwal M, Laszik ZG, Putnam AL, Lee K, Leung J, Nguyen V, Sigdel T, Tavares EC, Yang JY, Hellerstein M, Fitch M, Bluestone JA, Vincenti F. Polyclonal Regulatory T Cell Therapy for Control of Inflammation in Kidney Transplants. Am J Transplant 2017; 17:2945-2954. [PMID: 28675676 PMCID: PMC5662482 DOI: 10.1111/ajt.14415] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/23/2017] [Accepted: 06/25/2017] [Indexed: 01/25/2023]
Abstract
Early subclinical inflammation in kidney transplants is associated with later graft fibrosis and dysfunction. Regulatory T cells (Tregs) can reverse established inflammation in animal models. We conducted a pilot safety and feasibility trial of autologous Treg cell therapy in three kidney transplant recipients with subclinical inflammation noted on 6-month surveillance biopsies. Tregs were purified from peripheral blood and polyclonally expanded ex vivo using medium containing deuterated glucose to label the cells. All patients received a single infusion of ~320 × 106 (319, 321, and 363.8 × 106 ) expanded Tregs. Persistence of the infused Tregs was tracked. Graft inflammation was monitored with follow-up biopsies and urinary biomarkers. Nearly 1 × 109 (0.932, 0.956, 1.565 × 109 ) Tregs were successfully manufactured for each patient. There were no infusion reactions or serious therapy-related adverse events. The infused cells demonstrated patterns of persistence and stability similar to those observed in non-immunosuppressed subjects receiving the same dose of Tregs. Isolation and expansion of Tregs is feasible in kidney transplant patients on immunosuppression. Infusion of these cells was safe and well tolerated. Future trials will test the efficacy of polyclonal and donor alloantigen-reactive Tregs for the treatment of inflammation in kidney transplants.
Collapse
Affiliation(s)
- Sindhu Chandran
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, CA 94143, USA,Diabetes Center, University of California, San Francisco, CA 94143, USA
| | - Minnie Sarwal
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Zoltan G. Laszik
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Amy L. Putnam
- Diabetes Center, University of California, San Francisco, CA 94143, USA
| | - Karim Lee
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Joey Leung
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Vinh Nguyen
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Tara Sigdel
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Erica C. Tavares
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Joshua Y.C. Yang
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Marc Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Mark Fitch
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | | | - Flavio Vincenti
- Department of Medicine, University of California, San Francisco, CA 94143, USA,Department of Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
173
|
Ugor E, Simon D, Almanzar G, Pap R, Najbauer J, Németh P, Balogh P, Prelog M, Czirják L, Berki T. Increased proportions of functionally impaired regulatory T cell subsets in systemic sclerosis. Clin Immunol 2017; 184:54-62. [DOI: 10.1016/j.clim.2017.05.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/03/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022]
|
174
|
Tian Y, Chen T, Wu Y, Yang L, Wang L, Fan X, Zhang W, Feng J, Yu H, Yang Y, Zhou J, Yuan Z, Wu Y. Pioglitazone stabilizes atherosclerotic plaque by regulating the Th17/Treg balance in AMPK-dependent mechanisms. Cardiovasc Diabetol 2017; 16:140. [PMID: 29084546 PMCID: PMC5663071 DOI: 10.1186/s12933-017-0623-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/18/2017] [Indexed: 11/23/2022] Open
Abstract
Background Pioglitazone (PIO), a thiazolidinediones drug, is a well-known anti-diabetic medicine, but its anti-atherosclerotic effects remain controversial. Thus it is important to investigate the effects of PIO on atherogenesis and the relevant mechanisms. Methods For in vitro studies, primary cultured or AMP-activated protein kinase (AMPK) inhibited splenocytes were treated with oxidized low density lipoprotein (ox-LDL) or ox-LDL plus PIO. Percentage of T helper 17 (Th17) and regulatory T (Treg) cells were determined by flow cytometry. Expression of AMPK, interleukin-17 (IL-17) and forkhead box P3 (FoxP3) were detected by Western blots. For in vivo studies, apolipoprotein E–deficient (apoE−/−) mice fed with western diet were treated with PIO or vehicle for 8 weeks respectively. Percentage of Th17 and Treg cells in spleen were measured by immunohistochemical analysis. The atherosclerotic lesions were analyzed using oil red O staining, and collagen types I and III in atherosclerotic lesions were stained by Sirius red. Expression of IL-17 and FoxP3 were determined by quantitative polymerase chain reaction. Results In cultured primary splenocytes, PIO dramatically inhibited Th17 and raised Treg. Intriguingly, pharmacological and genetic AMPK inhibitions abolished PIO-induced Treg elevation and Th17 inhibition. Moreover, PIO significantly induced AMPK phosphorylation, decreased IL-17+ and increased FoxP3+ cells in spleen of apoE−/− mice. Finally, PIO did not alter plaque area, but intriguingly, stabilized atherosclerotic plaque through collagen induction in apoE−/− mice. PIO treatment also improved Th17/Treg balance in atherosclerotic lesions. Conclusions PIO exhibits anti-atherosclerotic effects for stabilization of atherosclerotic plaque through regulating the Th17/Treg balance in an AMPK-dependent manner.
Collapse
Affiliation(s)
- Yuling Tian
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Tao Chen
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yan Wu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Lin Yang
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Lijun Wang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xiaojuan Fan
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Wei Zhang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jiahao Feng
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Hang Yu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yanjie Yang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Juan Zhou
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Zuyi Yuan
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| | - Yue Wu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
175
|
García-González PA, Schinnerling K, Sepúlveda-Gutiérrez A, Maggi J, Mehdi AM, Nel HJ, Pesce B, Larrondo ML, Aravena O, Molina MC, Catalán D, Thomas R, Verdugo RA, Aguillón JC. Dexamethasone and Monophosphoryl Lipid A Induce a Distinctive Profile on Monocyte-Derived Dendritic Cells through Transcriptional Modulation of Genes Associated With Essential Processes of the Immune Response. Front Immunol 2017; 8:1350. [PMID: 29109727 PMCID: PMC5660598 DOI: 10.3389/fimmu.2017.01350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/03/2017] [Indexed: 02/02/2023] Open
Abstract
There is growing interest in the use of tolerogenic dendritic cells (tolDCs) as a potential target for immunotherapy. However, the molecular bases that drive the differentiation of monocyte-derived DCs (moDCs) toward a tolerogenic state are still poorly understood. Here, we studied the transcriptional profile of moDCs from healthy subjects, modulated with dexamethasone (Dex) and activated with monophosphoryl lipid A (MPLA), referred to as Dex-modulated and MPLA-activated DCs (DM-DCs), as an approach to identify molecular regulators and pathways associated with the induction of tolerogenic properties in tolDCs. We found that DM-DCs exhibit a distinctive transcriptional profile compared to untreated (DCs) and MPLA-matured DCs. Differentially expressed genes downregulated by DM included MMP12, CD1c, IL-1B, and FCER1A involved in DC maturation/inflammation and genes upregulated by DM included JAG1, MERTK, IL-10, and IDO1 involved in tolerance. Genes related to chemotactic responses, cell-to-cell signaling and interaction, fatty acid oxidation, metal homeostasis, and free radical scavenging were strongly enriched, predicting the activation of alternative metabolic processes than those driven by counterpart DCs. Furthermore, we identified a set of genes that were regulated exclusively by the combined action of Dex and MPLA, which are mainly involved in the control of zinc homeostasis and reactive oxygen species production. These data further support the important role of metabolic processes on the control of the DC-driven regulatory immune response. Thus, Dex and MPLA treatments modify gene expression in moDCs by inducing a particular transcriptional profile characterized by the activation of tolerance-associated genes and suppression of the expression of inflammatory genes, conferring the potential to exert regulatory functions and immune response modulation.
Collapse
Affiliation(s)
- Paulina A García-González
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Katina Schinnerling
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Alejandro Sepúlveda-Gutiérrez
- Programa de Genética Humana, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Jaxaira Maggi
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Ahmed M Mehdi
- Translational Research Institute, University of Queensland Diamantina Institute, Woolloongabba, QLD, Australia
| | - Hendrik J Nel
- Translational Research Institute, University of Queensland Diamantina Institute, Woolloongabba, QLD, Australia
| | - Bárbara Pesce
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Milton L Larrondo
- Banco de Sangre, Hospital Clínico de la Universidad de Chile, Santiago, Chile
| | - Octavio Aravena
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - María C Molina
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Diego Catalán
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Ranjeny Thomas
- Translational Research Institute, University of Queensland Diamantina Institute, Woolloongabba, QLD, Australia
| | - Ricardo A Verdugo
- Programa de Genética Humana, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Juan C Aguillón
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| |
Collapse
|
176
|
Li H, Chen HY, Liu WX, Jia XX, Zhang JG, Ma CL, Zhang XJ, Yu F, Cong B. Prostaglandin E 2 restrains human Treg cell differentiation via E prostanoid receptor 2-protein kinase A signaling. Immunol Lett 2017; 191:63-72. [PMID: 28963072 DOI: 10.1016/j.imlet.2017.09.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/01/2017] [Accepted: 09/25/2017] [Indexed: 10/18/2022]
Abstract
Regulatory T cells (Treg cells) belong to a class of immunosuppressive cells that control the pathological changes of autoimmunity and inflammation. Prostaglandin E2 (PGE2) is a potent lipid mediator of immune inflammation including rheumatoid arthritis (RA) that exerts its effects via four subtypes of G-protein-coupled receptors (EP1-4). The ability of PGE2 to regulate human Treg differentiation has not yet been reported. In the current study, we investigated the effects of PGE2 on the differentiation of naïve T cells from healthy and RA patients into Treg cells and the intracellular signaling involved in this process in vitro. Our data indicate that PGE2 negatively influenced the percentage of Treg cells and Foxp3 mRNA expression. The regulatory effects of PGE2 were associated with increased intracellular cAMP levels and PKA activity. EP2 receptors may mediate the inhibitory role of PGE2, since PGE2 actions were mimicked by EP2 agonist (Butaprost) and cAMP agonist (Sp-8-CPT-cAMPS) but were reversed by an EP2 antagonist (PF-04418948) and a PKA inhibitor (H-89). PGE2 negatively modulated the expression of cytotoxic T lymphocyte antigen-4 (CTLA-4) and glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR), as well as the production of interleukin (IL)-10 by Treg cells via EP2 receptors and cAMP/PKA signaling. All these findings indicate that PGE2 can inhibit Treg differentiation mediated through the EP2-cAMP/PKA signaling pathway, and suggest novel immune-based therapies for use in RA treatment.
Collapse
Affiliation(s)
- Hui Li
- Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Hai-Ying Chen
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050000, PR China.
| | - Wen-Xuan Liu
- Department of Forensic Medicine, Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Xian-Xian Jia
- Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Jing-Ge Zhang
- Department of Forensic Medicine, Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Chun-Ling Ma
- Department of Forensic Medicine, Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Xiao-Jing Zhang
- Department of Forensic Medicine, Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Feng Yu
- Department of Forensic Medicine, Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Bin Cong
- Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
177
|
NF-κB c-Rel Is Crucial for the Regulatory T Cell Immune Checkpoint in Cancer. Cell 2017; 170:1096-1108.e13. [PMID: 28886380 DOI: 10.1016/j.cell.2017.08.004] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/02/2017] [Accepted: 08/01/2017] [Indexed: 12/21/2022]
Abstract
Regulatory T cells (Tregs) play a pivotal role in the inhibition of anti-tumor immune responses. Understanding the mechanisms governing Treg homeostasis may therefore be important for development of effective tumor immunotherapy. We have recently demonstrated a key role for the canonical nuclear factor κB (NF-κB) subunits, p65 and c-Rel, in Treg identity and function. In this report, we show that NF-κB c-Rel ablation specifically impairs the generation and maintenance of the activated Treg (aTreg) subset, which is known to be enriched at sites of tumors. Using mouse models, we demonstrate that melanoma growth is drastically reduced in mice lacking c-Rel, but not p65, in Tregs. Moreover, chemical inhibition of c-Rel function delayed melanoma growth by impairing aTreg-mediated immunosuppression and potentiated the effects of anti-PD-1 immunotherapy. Our studies therefore establish inhibition of NF-κB c-Rel as a viable therapeutic approach for enhancing checkpoint-targeting immunotherapy protocols.
Collapse
|
178
|
Joshi RN, Binai NA, Marabita F, Sui Z, Altman A, Heck AJR, Tegnér J, Schmidt A. Phosphoproteomics Reveals Regulatory T Cell-Mediated DEF6 Dephosphorylation That Affects Cytokine Expression in Human Conventional T Cells. Front Immunol 2017; 8:1163. [PMID: 28993769 PMCID: PMC5622166 DOI: 10.3389/fimmu.2017.01163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/01/2017] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs) control key events of immune tolerance, primarily by suppression of effector T cells. We previously revealed that Tregs rapidly suppress T cell receptor (TCR)-induced calcium store depletion in conventional CD4+CD25− T cells (Tcons) independently of IP3 levels, consequently inhibiting NFAT signaling and effector cytokine expression. Here, we study Treg suppression mechanisms through unbiased phosphoproteomics of primary human Tcons upon TCR stimulation and Treg-mediated suppression, respectively. Tregs induced a state of overall decreased phosphorylation as opposed to TCR stimulation. We discovered novel phosphosites (T595_S597) in the DEF6 (SLAT) protein that were phosphorylated upon TCR stimulation and conversely dephosphorylated upon coculture with Tregs. Mutation of these DEF6 phosphosites abrogated interaction of DEF6 with the IP3 receptor and affected NFAT activation and cytokine transcription in primary Tcons. This novel mechanism and phosphoproteomics data resource may aid in modifying sensitivity of Tcons to Treg-mediated suppression in autoimmune disease or cancer.
Collapse
Affiliation(s)
- Rubin N Joshi
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Nadine A Binai
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Francesco Marabita
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Zhenhua Sui
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.,Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Angelika Schmidt
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
179
|
Differential regulation of Effector and Regulatory T cell function by Blimp1. Sci Rep 2017; 7:12078. [PMID: 28935958 PMCID: PMC5608714 DOI: 10.1038/s41598-017-12171-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/04/2017] [Indexed: 02/01/2023] Open
Abstract
The transcriptional regulator Blimp1 plays crucial roles in controlling terminal differentiation in several lineages. In T cells, Blimp1 is expressed in both effector (Teff) and regulatory (Treg) cells, and mice with T cell-specific deletion of Blimp1 (Blimp1CKO mice) spontaneously develop severe intestinal inflammation, indicating a crucial role for Blimp1 in T cell homeostasis regulation. Blimp1 has been shown to function as a direct activator of the Il10 gene and although its requirement for IL10 expression has been demonstrated in both Treg and Teff cells under inflammatory conditions, the intrinsic requirement of Blimp1 for homeostatic maintenance of these T cell subsets had not been investigated. Using mice with Foxp3+ Treg-cell specific deletion of Blimp1 and other approaches, here we show that Foxp3+ Treg cell-intrinsic expression of Blimp1 is required to control Treg and Teff cells homeostasis but, unexpectedly, it is dispensable to prevent development of severe spontaneous intestinal inflammation. In addition, we show that Blimp1 controls common and unique aspects of Treg and Teff cell function by differentially regulating gene expression in these T cell subsets. These findings document previously unappreciated aspects of Blimp1’s role in T cell biology and shed light on the intricate mechanisms regulating Treg and Teff cell function.
Collapse
|
180
|
Yang HY, Barbi J, Wu CY, Zheng Y, Vignali PDA, Wu X, Tao JH, Park BV, Bandara S, Novack L, Ni X, Yang X, Chang KY, Wu RC, Zhang J, Yang CW, Pardoll DM, Li H, Pan F. MicroRNA-17 Modulates Regulatory T Cell Function by Targeting Co-regulators of the Foxp3 Transcription Factor. Immunity 2017; 45:83-93. [PMID: 27438767 DOI: 10.1016/j.immuni.2016.06.022] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 12/16/2015] [Accepted: 02/24/2016] [Indexed: 01/06/2023]
Abstract
Regulatory T (Treg) cells are important in maintaining self-tolerance and immune homeostasis. The Treg cell transcription factor Foxp3 works in concert with other co-regulatory molecules, including Eos, to determine the transcriptional signature and characteristic suppressive phenotype of Treg cells. Here, we report that the inflammatory cytokine interleukin-6 (IL-6) actively repressed Eos expression through microRNA-17 (miR-17). miR-17 expression increased in Treg cells in the presence of IL-6, and its expression negatively correlated with that of Eos. Treg cell suppressive activity was diminished upon overexpression of miR-17 in vitro and in vivo, which was mitigated upon co-expression of an Eos mutant lacking miR-17 target sites. Also, RNAi of miR-17 resulted in enhanced suppressive activity. Ectopic expression of miR-17 imparted effector-T-cell-like characteristics to Treg cells via the de-repression of genes encoding effector cytokines. Thus, miR-17 provides a potent layer of Treg cell control through targeting Eos and additional Foxp3 co-regulators.
Collapse
Affiliation(s)
- Huang-Yu Yang
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Kidney Research Center, Department of Nephrology, Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Joseph Barbi
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Chao-Yi Wu
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Ying Zheng
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Department of Otolaryngology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Paolo D A Vignali
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xingmei Wu
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Department of Otolaryngology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Jin-Hui Tao
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Benjamin V Park
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Shashika Bandara
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lewis Novack
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xuhao Ni
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xiaoping Yang
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kwang-Yu Chang
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; National Institute of Cancer Research, NIH, Tainan 70456, Taiwan
| | - Ren-Chin Wu
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Junran Zhang
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Chih-Wei Yang
- Kidney Research Center, Department of Nephrology, Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Drew M Pardoll
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Huabin Li
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Department of Otolaryngology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| | - Fan Pan
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
181
|
Skuljec J, Chmielewski M, Happle C, Habener A, Busse M, Abken H, Hansen G. Chimeric Antigen Receptor-Redirected Regulatory T Cells Suppress Experimental Allergic Airway Inflammation, a Model of Asthma. Front Immunol 2017; 8:1125. [PMID: 28955341 PMCID: PMC5600908 DOI: 10.3389/fimmu.2017.01125] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 08/28/2017] [Indexed: 01/31/2023] Open
Abstract
Cellular therapy with chimeric antigen receptor (CAR)-redirected cytotoxic T cells has shown impressive efficacy in the treatment of hematologic malignancies. We explored a regulatory T cell (Treg)-based therapy in the treatment of allergic airway inflammation, a model for asthma, which is characterized by an airway hyper-reactivity (AHR) and a chronic, T helper-2 (Th2) cell-dominated immune response to allergen. To restore the immune balance in the lung, we redirected Tregs by a CAR toward lung epithelia in mice upon experimentally induced allergic asthma, closely mimicking the clinical situation. Adoptively transferred CAR Tregs accumulated in the lung and in tracheobronchial lymph nodes, reduced AHR and diminished eosinophilic airway inflammation, indicated by lower cell numbers in the bronchoalveolar lavage fluid and decreased cell infiltrates in the lung. CAR Treg cells furthermore prevented excessive pulmonary mucus production as well as increase in allergen-specific IgE and Th2 cytokine levels in exposed animals. CAR Tregs were more efficient in controlling asthma than non-modified Tregs, indicating the pivotal role of specific Treg cell activation in the affected organ. Data demonstrate that lung targeting CAR Treg cells ameliorate key features of experimental airway inflammation, paving the way for cell therapy of severe allergic asthma.
Collapse
Affiliation(s)
- Jelena Skuljec
- Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Markus Chmielewski
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Clinic I Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Christine Happle
- Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Anika Habener
- Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Mandy Busse
- Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Clinic I Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Gesine Hansen
- Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
182
|
Igarashi K, Kurosaki T, Roychoudhuri R. BACH transcription factors in innate and adaptive immunity. Nat Rev Immunol 2017; 17:437-450. [PMID: 28461702 DOI: 10.1038/nri.2017.26] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BTB and CNC homology (BACH) proteins are transcriptional repressors of the basic region leucine zipper (bZIP) transcription factor family. Recent studies indicate widespread roles of BACH proteins in controlling the development and function of the innate and adaptive immune systems, including the differentiation of effector and memory cells of the B and T cell lineages, CD4+ regulatory T cells and macrophages. Here, we emphasize similarities at a molecular level in the cell-type-specific activities of BACH factors, proposing that competitive interactions of BACH proteins with transcriptional activators of the bZIP family form a common mechanistic theme underlying their diverse actions. The findings contribute to a general understanding of how transcriptional repressors shape lineage commitment and cell-type-specific functions through repression of alternative lineage programmes.
Collapse
Affiliation(s)
- Kazuhiko Igarashi
- Department of Biochemistry, Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Tomohiro Kurosaki
- Laboratory for Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi-ku, Yokohama 230-0045, Japan
| | - Rahul Roychoudhuri
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK
| |
Collapse
|
183
|
Abstract
Modern immunosuppression regimens effectively control acute rejection and decrease graft loss in the first year after transplantation; however, these regimens do not have a durable effect on long-term graft survival owing to a combination of drug toxicities and the emergence of chronic alloimmune responses. Eliminating drugs and their toxicities while maintaining graft acceptance has been the primary aim of cellular therapies. Tregs suppress both autoimmune and alloimmune responses and are particularly effective in protecting allografts in experimental transplant models. Further, Treg-based therapies are selective, do not require harsh conditioning, and do not have a risk of graft-versus-host disease. Trial designs should consider the distinct immunological features of each transplanted organ, Treg preparations, dose, and frequency, and the ability to detect and quantify Treg effects in a given transplant environment. In this Review, we detail the ongoing clinical trials of Treg therapy in liver and kidney transplantation. Integration of Treg biology gleaned from preclinical models and experiences in human organ transplantation should allow for optimization of trial design that will determine the potential efficacy of a given therapy and provide guidelines for further therapeutic development.
Collapse
Affiliation(s)
- Qizhi Tang
- Department of Surgery.,Diabetes Center, and
| | - Flavio Vincenti
- Department of Surgery.,Department of Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
184
|
Ikeguchi R, Shimizu Y, Shimizu S, Kitagawa K. CSF and clinical data are useful in differentiating CNS inflammatory demyelinating disease from CNS lymphoma. Mult Scler 2017; 24:1212-1223. [DOI: 10.1177/1352458517717804] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: It is often difficult to diagnose central nervous system (CNS) inflammatory demyelinating diseases (IDDs) because they are similar to CNS lymphoma and glioma. Objective: To evaluate whether cerebrospinal fluid (CSF) analysis can differentiate CNS IDDs from CNS lymphoma and glioma. Methods: We measured CSF cell counts; concentrations of proteins, glucose, interleukin (IL)-6, IL-10, soluble IL-2 receptor (sIL-2R), and myelin basic protein; and IgG index in patients with multiple sclerosis (MS, n = 64), neuromyelitis optica spectrum disorder (NMOSD, n = 35), tumefactive demyelinating lesion (TDL, n = 17), CNS lymphoma ( n = 12), or glioma ( n = 10). We detected diagnostic markers using logistic regression and receiver operating characteristic (ROC) analyses. Results: Median CSF IL-10 and sIL-2R levels were higher in CNS lymphoma patients than in MS, NMOSD, or TDL patients. Logistic regression revealed that CSF sIL-2R levels predicted CNS lymphoma. In the ROC analysis of CSF sIL-2R levels, the area under the curve was 0.867, and the sensitivity and specificity were 83.3% and 90.0%, respectively. Conclusion: CSF sIL-2R levels can be used to differentiate CNS lymphoma from CNS IDDs. Further studies may identify other applications of CSF as a diagnostic biomarker.
Collapse
Affiliation(s)
- Ryotaro Ikeguchi
- Department of Neurology, School of Medicine, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yuko Shimizu
- Department of Neurology, School of Medicine, Tokyo Women’s Medical University, Tokyo, Japan
| | - Satoru Shimizu
- Medical Research Institute, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kazuo Kitagawa
- Department of Neurology, School of Medicine, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
185
|
Bansal SS, Ismahil MA, Goel M, Patel B, Hamid T, Rokosh G, Prabhu SD. Activated T Lymphocytes are Essential Drivers of Pathological Remodeling in Ischemic Heart Failure. Circ Heart Fail 2017; 10:e003688. [PMID: 28242779 DOI: 10.1161/circheartfailure.116.003688] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 01/23/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Inappropriately sustained inflammation is a hallmark of chronic ischemic heart failure (HF); however, the pathophysiological role of T lymphocytes is unclear. METHODS AND RESULTS Permanent coronary ligation was performed in adult C57BL/6 mice. When compared with sham-operated mice, mice with HF (8 weeks after ligation) exhibited the following features: (1) significant (P<0.05) expansion of circulating CD3+CD8+ cytotoxic and CD3+CD4+ helper (Th) T lymphocytes, together with increased Th1, Th2, Th17, and regulatory T-cell (Treg) CD4+ subsets; (2) significant expansion of CD8+ and CD4+ T cells in failing myocardium, with increased Th1, Th2, Th17, and Treg CD4+ subsets, marked reduction of the Th1/Th2 ratio, augmentation of the Th17/Treg ratio, and upregulation of Th2 cytokines; and (3) significantly increased Th1, Th2, Th17 cells, and Tregs, in the spleen and mediastinal lymph nodes, with expansion of splenic antigen-experienced effector and memory CD4+ T cells. Antibody-mediated CD4+ T-cell depletion in HF mice (starting 4 weeks after ligation) reduced cardiac infiltration of CD4+ T cells and prevented progressive left ventricular dilatation and hypertrophy, whereas adoptive transfer of splenic CD4+ T cells (and, to a lesser extent, cardiac CD3+ T cells) from donor mice with HF induced long-term left ventricular dysfunction, fibrosis, and hypertrophy in naive recipient mice. CONCLUSIONS CD4+ T lymphocytes are globally expanded and activated in chronic ischemic HF, with Th2 (versus Th1) and Th17 (versus Treg) predominance in failing hearts, and with expansion of memory T cells in the spleen. Cardiac and splenic T cells in HF are primed to induce cardiac injury and remodeling, and retain this memory on adoptive transfer.
Collapse
Affiliation(s)
- Shyam S Bansal
- From the Division of Cardiovascular Disease, Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., B.P., T.H., S.D.P.); and Medical Service, Birmingham Veterans Administration Medical Center, AL (S.S.B., S.D.P.)
| | - Mohamed Ameen Ismahil
- From the Division of Cardiovascular Disease, Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., B.P., T.H., S.D.P.); and Medical Service, Birmingham Veterans Administration Medical Center, AL (S.S.B., S.D.P.)
| | - Mehak Goel
- From the Division of Cardiovascular Disease, Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., B.P., T.H., S.D.P.); and Medical Service, Birmingham Veterans Administration Medical Center, AL (S.S.B., S.D.P.)
| | - Bindiya Patel
- From the Division of Cardiovascular Disease, Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., B.P., T.H., S.D.P.); and Medical Service, Birmingham Veterans Administration Medical Center, AL (S.S.B., S.D.P.)
| | - Tariq Hamid
- From the Division of Cardiovascular Disease, Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., B.P., T.H., S.D.P.); and Medical Service, Birmingham Veterans Administration Medical Center, AL (S.S.B., S.D.P.)
| | - Gregg Rokosh
- From the Division of Cardiovascular Disease, Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., B.P., T.H., S.D.P.); and Medical Service, Birmingham Veterans Administration Medical Center, AL (S.S.B., S.D.P.)
| | - Sumanth D Prabhu
- From the Division of Cardiovascular Disease, Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., B.P., T.H., S.D.P.); and Medical Service, Birmingham Veterans Administration Medical Center, AL (S.S.B., S.D.P.).
| |
Collapse
|
186
|
Sang LX, Chang B, Zhu JF, Yang FL, Li Y, Jiang XF, Wang DN, Lu CL, Sun X. Sodium selenite ameliorates dextran sulfate sodium-induced chronic colitis in mice by decreasing Th1, Th17, and γδT and increasing CD4(+)CD25(+) regulatory T-cell responses. World J Gastroenterol 2017; 23:3850-3863. [PMID: 28638225 PMCID: PMC5467071 DOI: 10.3748/wjg.v23.i21.3850] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/29/2016] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the effect of sodium selenite on the severity of dextran sulfate sodium (DSS)-induced colitis in C57BL/6 mice.
METHODS Mice were randomly divided into four groups (n = 10/group): normal group, selenium (Se) group, chronic colitis group, and Se + chronic colitis group. The mice were sacrificed on day 26. Survival rates, clinical symptoms, colon length, and histological changes were determined. The percentages and absolute numbers of immune system cells in the lamina propria lymphocytes (LPL) of the colon, the expression of mRNA in colon tissue, and the concentrations of Th1, Th17, and Treg cytokines in LPL from the large intestine, were measured.
RESULTS Se significantly ameliorated the symptoms of colitis and histological injury (P < 0.05 each), increasing the proportions of neutrophils and CD4+ CD25+ T cells (P < 0.05 each) and decreasing the proportions of γδT cells, CD4+, CD4+CD44+, and CD4+ CD69+ T cells in LPL (P < 0.05 each). Moreover, Se reduced the expression of IL-6, IFN-γ, IL-17A, IL-21, T-bet, and RORγt (P < 0.05 each), but enhanced the expression of IL-10 and Foxp3 (P < 0.05 each).
CONCLUSION These results suggest that Se protects against DSS-induced chronic colitis perhaps by increasing the number of CD4(+)CD25(+) Tregs that suppress the secretion of proinflammatory cytokines and populations of Th1, Th17, and γδT cells.
Collapse
|
187
|
Badal D, Kumar R, Paul M, Dayal D, Bhansali A, Bhadada SK, Kumar R, Sachdeva N. Peripheral blood mononuclear cells of patients with latent autoimmune diabetes secrete higher levels of pro- & anti-inflammatory cytokines compared to those with type-1 diabetes mellitus following in vitro stimulation with β-cell autoantigens. Indian J Med Res 2017; 145:767-776. [PMID: 29067979 PMCID: PMC5674547 DOI: 10.4103/ijmr.ijmr_1563_15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND & OBJECTIVES Type-1 diabetes mellitus (T1DM) and latent autoimmune diabetes in adults (LADA) share similar pathological features but differ in age of onset and progression. There is a scarcity of information on differences in CD4+ T-cell responses, particularly, cytokine secretion, between the two forms of autoimmune diabetes. Here proliferative potential and concentration of pro- and anti-inflammatory cytokines secreted by peripheral blood mononuclear cells (PBMCs) of T1DM and LADA patients were compared, after in vitro stimulation with β-cell autoantigens. METHODS A total of 19 patients with LADA, 37 with T1DM and 20 healthy controls were compared on the basis of lymphocyte proliferation and secretion of pro- and anti-inflammatory cytokines belonging to different T-helper types after in vitro stimulation of PBMCs with insulin and glutamic acid decarboxylase 65 (GAD65). RESULTS Following insulin stimulation, LADA group secreted higher concentration of interleukin-17 (IL-17) (P=0.02) and had higher proportion of interferon gamma (IFN-γ) secretors (P<0.001) than T1DM group. Post-GAD65 stimulation, higher proportion of LADA patients secreted IL-23 than T1DM group (P=0.02). Proportion of responders , as well as levels of secreted IL-10, were significantly higher in LADA than T1DM group, following stimulation with both insulin (P=0.01) and GAD65 (P=0.03). A significant positive correlation was observed between body mass index and IL-17 levels (r=0.41, P=0.04) and fasting plasma C-peptide with IL-10 levels (r=0.37, P=0.04). INTERPRETATION & CONCLUSIONS There are differences in the portfolio of cytokine secretion in diabetic subjects with varying rates of β-cell destruction as LADA subjects secrete higher levels of both pro- and anti-inflammatory cytokines on exposure to β-cell autoantigens, thus highlighting another distinguishing feature in the pathophysiology of the two forms of autoimmune diabetes.
Collapse
Affiliation(s)
- Darshan Badal
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Rajendra Kumar
- University Institute of Pharmaceutical Sciences (UIPS), Chandigarh, India
| | - Mahinder Paul
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Devi Dayal
- Department of Pediatrics, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Anil Bhansali
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Sanjay Kumar Bhadada
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Rajesh Kumar
- Department of Statistics, Panjab University, Chandigarh, India
| | - Naresh Sachdeva
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
188
|
Tselios K, Sarantopoulos A, Gkougkourelas I, Boura P. T Regulatory Cells in Systemic Lupus Erythematosus: Current Knowledge and Future Prospects. Lupus 2017. [DOI: 10.5772/intechopen.68479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
189
|
Baekkeskov S, Hubbell JA, Phelps EA. Bioengineering strategies for inducing tolerance in autoimmune diabetes. Adv Drug Deliv Rev 2017. [PMID: 28625830 DOI: 10.1016/j.addr.2017.06.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes is an autoimmune disease marked by the destruction of insulin-producing beta cells in the pancreatic islets. Strategies to delay onset or prevent the autoimmune recognition of beta cell antigens or T cell-mediated killing of beta cells have mainly focused on systemic immunomodulation and antigen-specific immunotherapy. To bridge the fields of type 1 diabetes immunology and biomaterials engineering, this article will review recent trends in the etiology of type 1 diabetes immunopathology and will focus on the contributions of emerging bioengineered strategies in the fight against beta cell autoimmunity in type 1 diabetes.
Collapse
Affiliation(s)
- Steinunn Baekkeskov
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Building SV 3826, Station 19, CH-1015 Lausanne, Switzerland; Departments of Medicine and Microbiology/Immunology, Diabetes Center, 513 Parnassus Ave, 20159, Box 0534, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jeffrey A Hubbell
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Building SV 3826, Station 19, CH-1015 Lausanne, Switzerland; Institute for Molecular Engineering, University of Chicago, 5640 S Ellis Avenue, Chicago, IL 60615, USA
| | - Edward A Phelps
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Building SV 3826, Station 19, CH-1015 Lausanne, Switzerland; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, P.O. Box 116131, Gainesville, FL 32611, USA.
| |
Collapse
|
190
|
Effector and Regulatory T Cell Trafficking in Corneal Allograft Rejection. Mediators Inflamm 2017; 2017:8670280. [PMID: 28539707 PMCID: PMC5429952 DOI: 10.1155/2017/8670280] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/26/2017] [Indexed: 01/08/2023] Open
Abstract
Corneal transplantation is among the most prevalent and successful forms of solid tissue transplantation in humans. Failure of corneal allograft is mainly due to immune-mediated destruction of the graft, a complex and highly coordinated process that involves elaborate interactions between cells of innate and adaptive immunity. The migration of immune cells to regional lymphoid tissues and to the site of graft plays a central role in the immunopathogenesis of graft rejection. Intricate interactions between adhesion molecules and their counter receptors on immune cells in conjunction with tissue-specific chemokines guide the trafficking of these cells to the draining lymph nodes and ultimately to the site of graft. In this review, we discuss the cascade of chemokines and adhesion molecules that mediate the trafficking of effector and regulatory T cells during corneal allograft rejection.
Collapse
|
191
|
Walker LS. EFIS Lecture: Understanding the CTLA-4 checkpoint in the maintenance of immune homeostasis. Immunol Lett 2017; 184:43-50. [DOI: 10.1016/j.imlet.2017.02.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 02/13/2017] [Indexed: 01/08/2023]
|
192
|
Lee HJ, Kim SN, Jeon MS, Yi T, Song SU. ICOSL expression in human bone marrow-derived mesenchymal stem cells promotes induction of regulatory T cells. Sci Rep 2017; 7:44486. [PMID: 28290526 PMCID: PMC5349520 DOI: 10.1038/srep44486] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 02/09/2017] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can modulate lymphocyte proliferation and function. One of the immunomodulatory functions of MSCs involves CD4+CD25+FoxP3+ regulatory T cells (Tregs), which negatively regulate inflammatory responses. MSC-mediated Treg induction is supposed to be regulated by mechanisms requiring both soluble and cell contact-dependent factors. Although the involvement of soluble factors has been revealed, the contact-dependent mechanisms in MSC-mediated Treg induction remain unclear. We attempted to identify molecule(s) other than secreted factors that are responsible for MSC-mediated Treg induction and to uncover the underlying mechanisms. Under in vitro Treg-inducing conditions, ICOSL expression in MSCs coincided with Treg induction in co-cultures of MSCs with CD4+ T cells. When cultured in a transwell plate, MSCs failed to induce Tregs. Neutralization or knockdown of ICOSL significantly reduced Tregs and their IL-10 release. ICOSL overexpression in MSCs promoted induction of functional Tregs. ICOSL-ICOS signaling promoted Treg differentiation from CD4+ T cells through activation of the phosphoinositide 3-kinase-Akt pathway. MSCs primed with Interleukin-1β significantly induced Tregs through ICOSL upregulation. We demonstrated that the Treg-inducing activity of MSCs is proportionate to their basal ICOSL expression. This study provides evidence that ICOSL expression in human MSCs plays an important role in contact-dependent regulation of MSC-mediated Treg induction.
Collapse
Affiliation(s)
- Hyun-Joo Lee
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea.,Drug Development Program, Department of Biomedical Sciences, Inha University School of Medicine, Incheon, Republic of Korea
| | - Si-Na Kim
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea.,Drug Development Program, Department of Biomedical Sciences, Inha University School of Medicine, Incheon, Republic of Korea.,SCM Lifesciences Co. Ltd., Incheon, Republic of Korea
| | - Myung-Shin Jeon
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea
| | - TacGhee Yi
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea.,SunCreate Co. Ltd., Yangju, Republic of Korea
| | - Sun U Song
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea.,SCM Lifesciences Co. Ltd., Incheon, Republic of Korea
| |
Collapse
|
193
|
Green AR, Aleskandarany MA, Ali R, Hodgson EG, Atabani S, De Souza K, Rakha EA, Ellis IO, Madhusudan S. Clinical Impact of Tumor DNA Repair Expression and T-cell Infiltration in Breast Cancers. Cancer Immunol Res 2017; 5:292-299. [PMID: 28254786 DOI: 10.1158/2326-6066.cir-16-0195] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/22/2016] [Accepted: 02/20/2017] [Indexed: 11/16/2022]
Abstract
Impaired DNA repair drives mutagenicity, which increases neoantigen load and immunogenicity. We investigated the expression of proteins involved in the DNA damage response (ATM, Chk2), double-strand break repair (BRCA1, BLM, WRN, RECQL4, RECQL5, TOPO2A, DNA-PKcs, Ku70/Ku80), nucleotide excision repair (ERCC1), base excision repair (XRCC1, pol β, FEN1, PARP1), and immune responses (CD8, PD-1, PD-L1, FOXP3) in 1,269 breast cancers and validated our findings in an independent estrogen receptor-negative (ER-) cohort (n = 279). Patients with tumors that expressed low XRCC1, low ATM, and low BRCA1 were not only associated with high numbers of CD8+ tumor-infiltrating lymphocytes, but were also linked to higher grades, high proliferation indexes, presence of dedifferentiated cells, ER- cells, and poor survival (all P ≤ 0.01). PD-1+ or PD-L1+ breast cancers with low XRCC1 were also linked to an aggressive phenotype that was high grade, had high proliferation indexes, contained dedifferentiated cells and ER- (all with P values ≤ 0.01), and poor survival (P = 0.00021 and P = 0.00022, for PD-1+ and PD-L1+ cancers, respectively) including in an independent ER- validation cohort (P = 0.007 and P = 0.047, respectively). We conclude that the interplay between DNA repair, CD8, PD-L1, and PD-1 can promote aggressive tumor phenotypes. XRCC1-directed personalization of immune checkpoint inhibitor therapy may be feasible and warrants further investigation in breast cancer. Cancer Immunol Res; 5(4); 292-9. ©2017 AACR.
Collapse
Affiliation(s)
- Andrew R Green
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Mohammed A Aleskandarany
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Reem Ali
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Eleanor Grace Hodgson
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Suha Atabani
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Karen De Souza
- Department of Oncology, Nottingham University Hospitals, Nottingham, United Kingdom
| | - Emad A Rakha
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Ian O Ellis
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Srinivasan Madhusudan
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom. .,Department of Oncology, Nottingham University Hospitals, Nottingham, United Kingdom
| |
Collapse
|
194
|
Ramjee V, Li D, Manderfield LJ, Liu F, Engleka KA, Aghajanian H, Rodell CB, Lu W, Ho V, Wang T, Li L, Singh A, Cibi DM, Burdick JA, Singh MK, Jain R, Epstein JA. Epicardial YAP/TAZ orchestrate an immunosuppressive response following myocardial infarction. J Clin Invest 2017; 127:899-911. [PMID: 28165342 DOI: 10.1172/jci88759] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/12/2016] [Indexed: 12/27/2022] Open
Abstract
Ischemic heart disease resulting from myocardial infarction (MI) is the most prevalent form of heart disease in the United States. Post-MI cardiac remodeling is a multifaceted process that includes activation of fibroblasts and a complex immune response. T-regulatory cells (Tregs), a subset of CD4+ T cells, have been shown to suppress the innate and adaptive immune response and limit deleterious remodeling following myocardial injury. However, the mechanisms by which injured myocardium recruits suppressive immune cells remain largely unknown. Here, we have shown a role for Hippo signaling in the epicardium in suppressing the post-infarct inflammatory response through recruitment of Tregs. Mice deficient in epicardial YAP and TAZ, two core Hippo pathway effectors, developed profound post-MI pericardial inflammation and myocardial fibrosis, resulting in cardiomyopathy and death. Mutant mice exhibited fewer suppressive Tregs in the injured myocardium and decreased expression of the gene encoding IFN-γ, a known Treg inducer. Furthermore, controlled local delivery of IFN-γ following MI rescued Treg infiltration into the injured myocardium of YAP/TAZ mutants and decreased fibrosis. Collectively, these results suggest that epicardial Hippo signaling plays a key role in adaptive immune regulation during the post-MI recovery phase.
Collapse
|
195
|
Mahmoudian-Sani MR, Asadi-Samani M, Luther T, Saeedi-Boroujeni A, Gholamian N. A new approach for treatment of type 1 diabetes: Phytotherapy and phytopharmacology of regulatory T cells. J Renal Inj Prev 2017. [DOI: 10.15171/jrip.2017.31] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
196
|
Lagatie O, Van Dorst B, Stuyver LJ. Identification of three immunodominant motifs with atypical isotype profile scattered over the Onchocerca volvulus proteome. PLoS Negl Trop Dis 2017; 11:e0005330. [PMID: 28125577 PMCID: PMC5295699 DOI: 10.1371/journal.pntd.0005330] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/07/2017] [Accepted: 01/14/2017] [Indexed: 01/30/2023] Open
Abstract
Understanding the immune response upon infection with the filarial nematode Onchocerca volvulus and the mechanisms that evolved in this parasite to evade immune mediated elimination is essential to expand the toolbox available for diagnostics, therapeutics and vaccines development. Using high-density peptide microarrays we scanned the proteome-wide linear epitope repertoire in Cameroonian onchocerciasis patients and healthy controls from Southern Africa which led to the identification of 249 immunodominant antigenic peptides. Motif analysis learned that 3 immunodominant motifs, encompassing 3 linear epitopes, are present in 70, 43, and 31 of these peptides, respectively and appear to be scattered over the entire proteome in seemingly non-related proteins. These linear epitopes are shown to have an atypical isotype profile dominated by IgG1, IgG3, IgE and IgM, in contrast to the commonly observed IgG4 response in chronic active helminth infections. The identification of these linear epitope motifs may lead to novel diagnostic development but further evaluation of cross-reactivity against common co-infecting human nematode infections will be needed. Infection with the filarial parasite Onchocerca volvulus is the cause of river blindness. We analyzed the immune response against this parasite in infected individuals in order to identify linear epitopes. Using high-density peptide microarrays we discovered three immunodominant motifs in the Onchocerca volvulus proteome that induce a broad IgG response, but the typical IgG4 immune response against parasites was absent. Our study led to the identification of novel potential epitope sequences that can potentially be used for detection of infection with Onchocerca volvulus.
Collapse
Affiliation(s)
- Ole Lagatie
- Janssen Diagnostics, Janssen Pharmaceutica NV, Beerse, Belgium
- * E-mail:
| | - Bieke Van Dorst
- Janssen Diagnostics, Janssen Pharmaceutica NV, Beerse, Belgium
| | | |
Collapse
|
197
|
Zhang H. Upregulation of PIM2 by Underexpression of MicroRNA-135-5p Improves Survival Rates of Skin Allografts by Suppressing Apoptosis of Fibroblast Cells. Med Sci Monit 2017; 23:107-113. [PMID: 28064305 PMCID: PMC5240881 DOI: 10.12659/msm.897613] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND It has been reported that miR-135-5p is involved with many diseases. In this study, we aimed at define the relationship between miR-135-5p level and burn patient survival after skin transplantation. MATERIAL AND METHODS Expression of miR-135-5p and PIM2 was measured using real-time PCR and Western blot analysis in the skin samples collected from burn patients who received skin graft or in the fibroblast cells transfected with miR-135-5p mimics or inhibitors. The regulatory association between miR-135-5p and PIM2 was verified using bioinformatics analysis and luciferase assay. RESULTS The expression level of miR-135-5p was determined in 60 tissue samples divided into 2 groups based on the presence of rejection (long survival n=30, and short survival n=30). We found that miR-135-5p was substantially downregulated in the long survival group. We then searched the miRNA database online with the "seed sequence" located within the 3'-UTR of the target gene, and then validated PIM2 to be the direct gene via luciferase reporter assay system. We also established the negative regulatory relationship between miR-135-5p and PIM2 via studying the relative luciferase activity. We also conducted real-time PCR and Western blot analysis to study the mRNA and protein expression level of PIM2 among different groups (long survival n=30, short survival n=30) or cells treated with scramble control, miR-135-5p mimics, PIM2 siRNA, and miR-135-5p inhibitors, indicating the negative regulatory relationship between MiR-135-5p and PIM2. We also conducted experiments to investigate the influence of miR-135-5p and PIM2 on viability and apoptosis of cells. The results showed miR-135-5p reduced the viability of cells, while PIM2 negatively interfered with the viability of cells, and miR-135-5p inhibited apoptosis and PIM2 suppressed apoptosis. CONCLUSIONS MiR-135-5p is involved with the prognosis of burn patients after skin transplantation. PIM2 is a virtual target of miR-135-5p, and there is a negative regulatory relationship between miR-135-5p and PIM2. MiR-135-5p and PIM2 interfered with the viability and apoptosis in cells.
Collapse
Affiliation(s)
- Hongtu Zhang
- Department of Burn and Plastic Surgery, Jining Number 1 People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|
198
|
Groeger S, Jarzina F, Domann E, Meyle J. Porphyromonas gingivalis activates NFκB and MAPK pathways in human oral epithelial cells. BMC Immunol 2017; 18:1. [PMID: 28056810 PMCID: PMC5217430 DOI: 10.1186/s12865-016-0185-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/16/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The bacterial biofilm at the gingival margin induces a host immune reaction. In this local inflammation epithelial cells defend the host against bacterial challenge. Porphyromonas gingivalis (P. gingivalis), a keystone pathogen, infects epithelial cells. The aim of this study was to investigate the activation of signaling cascades in primary epithelial cells and oral cancer cell lines by a profiler PCR array. RESULTS After infection with P. gingivalis membranes the RNA of 16 to 33 of 84 key genes involved in the antibacterial immune response was up-regulated, amongst them were IKBKB (NF-κB signaling pathway), IRF5 (TLR signaling) and JUN, MAP2K4, MAPK14 and MAPK8 (MAPK pathway) in SCC-25 cells and IKBKB, IRF5, JUN, MAP2K4, MAPK14 and MAPK8 in PHGK. Statistically significant up-regulation of IKBKB (4.7 ×), MAP2K4 (4.6 ×), MAPK14 (4.2 ×) and IRF5 (9.8 ×) (p < 0.01) was demonstrated in SCC-25 cells and IKBKB (3.1 ×), MAP2K4 (4.0 ×) MAPK 14 (3.0 ×) (p < 0.05), IRF5 (3.0 ×) and JUN (7.7 ×) (p < 0.01) were up-regulated in PHGK. CONCLUSIONS P. gingivalis membrane up-regulates the expression of genes involved in downstream TLR, NFκB and MAPK signaling pathways involved in the pro-inflammatory immune response in primary and malignant oral epithelial cells.
Collapse
Affiliation(s)
- Sabine Groeger
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany.
| | - Fabian Jarzina
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Eugen Domann
- Institute for Medical Microbiology - German Center for Infection Research, DZIF Partner Site Giessen-Marburg-Langen - Justus-Liebig-University of Giessen, Giessen, Germany
| | - Joerg Meyle
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
199
|
Waight JD, Gombos RB, Wilson NS. Harnessing co-stimulatory TNF receptors for cancer immunotherapy: Current approaches and future opportunities. Hum Antibodies 2017; 25:87-109. [PMID: 28085016 DOI: 10.3233/hab-160308] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Co-stimulatory tumor necrosis factor receptors (TNFRs) can sculpt the responsiveness of T cells recognizing tumor-associated antigens. For this reason, agonist antibodies targeting CD137, CD357, CD134 and CD27 have received considerable attention for their therapeutic utility in enhancing anti-tumor immune responses, particularly in combination with other immuno-modulatory antibodies targeting co-inhibitory pathways in T cells. The design of therapeutic antibodies that optimally engage and activate co-stimulatory TNFRs presents an important challenge of how to promote effective anti-tumor immunity while avoiding serious immune-related adverse events. Here we review our current understanding of the expression, signaling and structural features of CD137, CD357, CD134 and CD27, and how this may inform the design of pharmacologically active immuno-modulatory antibodies targeting these receptors. This includes the integration of our emerging knowledge of the role of Fcγ receptors (FcγRs) in facilitating antibody-mediated receptor clustering and forward signaling, as well as promoting immune effector cell-mediated activities. Finally, we bring our current preclinical and clinical knowledge of co-stimulatory TNFR antibodies into the context of opportunities for next generation molecules with improved pharmacologic properties.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- Gene Expression Regulation
- Humans
- Immunity, Cellular/drug effects
- Immunotherapy/methods
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Receptors, IgG/agonists
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Receptors, Tumor Necrosis Factor/agonists
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/immunology
- Signal Transduction
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
|
200
|
Abstract
Immunotherapy using dendritic cell (DC)-based vaccination is an approved approach for harnessing the potential of a patient's own immune system to eliminate tumor cells in metastatic hormone-refractory cancer. Overall, although many DC vaccines have been tested in the clinic and proven to be immunogenic, and in some cases associated with clinical outcome, there remains no consensus on how to manufacture DC vaccines. In this review we will discuss what has been learned thus far about human DC biology from clinical studies, and how current approaches to apply DC vaccines in the clinic could be improved to enhance anti-tumor immunity.
Collapse
|