1
|
Søndergaard JN, Tulyeu J, Priest D, Sakaguchi S, Wing JB. Single cell suppression profiling of human regulatory T cells. Nat Commun 2025; 16:1325. [PMID: 39900891 PMCID: PMC11791207 DOI: 10.1038/s41467-024-55746-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Regulatory T cells (Treg) play an important role in regulating immune homeostasis in health and disease. Traditionally their suppressive function has been assayed by mixing purified cell populations, which does not provide an accurate picture of a physiologically relevant response. To overcome this limitation, we here develop 'single cell suppression profiling of human Tregs' (scSPOT). scSPOT uses a 52-marker CyTOF panel, a cell division detection algorithm, and a whole PBMC system to assess the effect of Tregs on all other cell types simultaneously. In this head-to-head comparison, we find Tregs having the clearest suppressive effects on effector memory CD8 T cells through partial division arrest, cell cycle inhibition, and effector molecule downregulation. Additionally, scSPOT identifies a Treg phenotypic split previously observed in viral infection and propose modes of action by the FDA-approved drugs Ipilimumab and Tazemetostat. scSPOT is thus scalable, robust, widely applicable, and may be used to better understand Treg immunobiology and screen for therapeutic compounds.
Collapse
Affiliation(s)
- Jonas Nørskov Søndergaard
- Human Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan.
| | - Janyerkye Tulyeu
- Human Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - David Priest
- Laboratory of Human Single Cell Immunology, WPI-IFReC, Osaka University, Suita, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, WPI-IFReC, Osaka University, Suita, Japan
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - James B Wing
- Human Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan.
- Laboratory of Human Single Cell Immunology, WPI-IFReC, Osaka University, Suita, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan.
| |
Collapse
|
2
|
Lazarski CA, Hanley PJ. Review of flow cytometry as a tool for cell and gene therapy. Cytotherapy 2024; 26:103-112. [PMID: 37943204 PMCID: PMC10872958 DOI: 10.1016/j.jcyt.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Quality control testing and analytics are critical for the development and manufacture of cell and gene therapies, and flow cytometry is a key quality control and analytical assay that is used extensively. However, the technical scope of characterization assays and safety assays must keep apace as the breadth of cell therapy products continues to expand beyond hematopoietic stem cell products into producing novel adoptive immune therapies and gene therapy products. Flow cytometry services are uniquely positioned to support the evolving needs of cell therapy facilities, as access to flow cytometers, new antibody clones and improved fluorochrome reagents becomes more egalitarian. This report will outline the features, logistics, limitations and the current state of flow cytometry within the context of cellular therapy.
Collapse
Affiliation(s)
- Christopher A Lazarski
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| | - Patrick J Hanley
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| |
Collapse
|
3
|
Luo Y, Acevedo D, Vlagea A, Codina A, García-García A, Deyà-Martínez A, Martí-Castellote C, Esteve-Solé A, Alsina L. Changes in Treg and Breg cells in a healthy pediatric population. Front Immunol 2023; 14:1283981. [PMID: 38077340 PMCID: PMC10704817 DOI: 10.3389/fimmu.2023.1283981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/31/2023] [Indexed: 12/18/2023] Open
Abstract
The interpretation of clinical diagnostic results in suspected inborn errors of immunity, including Tregopathies, is hampered by the lack of age-stratified reference values for regulatory T cells (Treg) in the pediatric population and a consensus on which Treg immunophenotype to use. Regulatory B cells (Breg) are an important component of the regulatory system that have been poorly studied in the pediatric population. We analyzed (1) the correlation between the three immunophenotypic definitions of Treg (CD4+CD25hiCD127low, CD4+CD25hiCD127lowFoxP3+, CD4+CD25hiFoxP3+), and with CD4+CD25hi and (2) the changes in Treg and Breg frequencies and their maturation status with age. We performed peripheral blood immunophenotyping of Treg and Breg (CD19+CD24hiCD38hi) by flow cytometry in 55 healthy pediatric controls. We observed that Treg numbers varied depending on the definition used, and the frequency ranged between 3.3-9.7% for CD4+CD25hiCD127low, 0.07-1.6% for CD4+CD25hiCD127lowFoxP3+, and 0.24-2.83% for CD4+CD25hiFoxP3+. The correlation between the three definitions of Treg was positive for most age ranges, especially between the two intracellular panels and with CD4+CD25hi vs CD4+CD25hiCD127low. Treg and Breg frequencies tended to decline after 7 and 3 years onwards, respectively. Treg's maturation status increased with age, with a decline of naïve Treg and an increase in memory/effector Treg from age 7 onwards. Memory Breg increased progressively from age 3 onwards. In conclusion, the number of Treg frequencies spans a wide range depending on the immunophenotypic definition used despite a good level of correlation exists between them. The decline in numbers and maturation process with age occurs earlier in Breg than in Treg.
Collapse
Affiliation(s)
- Yiyi Luo
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Daniel Acevedo
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Alexandru Vlagea
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Biomedic Diagnostic Center (CDB), Hospital Clínic of Barcelona, Clinical Immunology Unit Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain
| | - Anna Codina
- Biobanco Pediátrico para la Investigación Hospital Sant Joan de Déu, Barcelona, Spain
| | - Ana García-García
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Angela Deyà-Martínez
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Celia Martí-Castellote
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Ana Esteve-Solé
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Department of Surgery and Medical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Ganesan N, Ronsmans S, Hoet P. Comparing [ 3H] thymidine LPT and CFSE assay to assess lymphocyte proliferation in beryllium-exposed sarcoidosis patients. Heliyon 2023; 9:e19242. [PMID: 37662805 PMCID: PMC10471999 DOI: 10.1016/j.heliyon.2023.e19242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/27/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
The detection of antigen specific lymphocyte responses plays a vital role in the diagnosis of various diseases. Beryllium-specific [3H] thymidine lymphocyte proliferation test (LPT) is regarded as a gold standard in identifying chronic beryllium disease (CBD) cases. Alternatively, flow cytometric based carboxyfluorescein succinimidyl ester (CFSE) assay, has several benefits as opposed to LPT, since it further permits both phenotypical characterization and functional analysis of proliferating lymphocyte subsets. The suitability of both LPT and CFSE assay to therefore detect beryllium sensitivity in a group of Be-exposed sarcoidosis patients with suspected beryllium exposure, was evaluated in this study. The clinical relevance of the test responses, expressed as stimulation indices (SI), were additionally compared on a group and individual level. Agreement in clinical interpretation of the test responses between both methods was observed in 4 out of 5 recruited patients, when considering total lymphocyte population i.e., CD3+ and CD19+-cells combined, on day 7 and with CFSE-SI >1.5, when compared with LPT-SI >2.5. Variability in responses to beryllium was additionally evaluated in Be-exposed sarcoidosis patients and compared with healthy controls. To conclude, both LPT and CFSE assay are suitable assays to detect Be sensitivity in Be-exposed sarcoidosis patients. At the same time, flow cytometric based CFSE assay has the edge over LPT in identifying the relevant proliferating lymphocyte populations. As such, when comparing two or more methods, factors that contribute to assay variability such as timepoints, lymphocyte subsets and number of replicates should always be accounted for.
Collapse
Affiliation(s)
- Nirosha Ganesan
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Steven Ronsmans
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Clinic for Occupational and Environmental Medicine, Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Peter Hoet
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Ganesan N, Ronsmans S, Hoet P. Methods to Assess Proliferation of Stimulated Human Lymphocytes In Vitro: A Narrative Review. Cells 2023; 12:cells12030386. [PMID: 36766728 PMCID: PMC9913443 DOI: 10.3390/cells12030386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/10/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
The ability to monitor lymphocyte responses is critical for developing our understanding of the immune response in humans. In the current clinical setting, relying on the metabolic incorporation of [3H] thymidine into cellular DNA via a lymphocyte proliferation test (LPT) is the only method that is routinely performed to determine cell proliferation. However, techniques that measure DNA synthesis with a radioactive material such as [3H] thymidine are intrinsically more sensitive to the different stages of the cell cycle, which could lead to over-analyses and the subsequent inaccurate interpretation of the information provided. With cell proliferation assays, the output should preferably provide a direct and accurate measurement of the number of actively dividing cells, regardless of the stimuli properties or length of exposure. In fact, an ideal technique should have the capacity to measure lymphocyte responses on both a quantitative level, i.e., cumulative magnitude of lymphoproliferative response, and a qualitative level, i.e., phenotypical and functional characterization of stimulated immune cells. There are many LPT alternatives currently available to measure various aspects of cell proliferation. Of the nine techniques discussed, we noted that the majority of these LPT alternatives measure lymphocyte proliferation using flow cytometry. Across some of these alternatives, the covalent labelling of cells with a high fluorescence intensity and low variance with minimal cell toxicity while maximizing the number of detectable cell divisions or magnitude of proliferation was achieved. Herein, we review the performance of these different LPT alternatives and address their compatibility with the [3H] thymidine LPT so as to identify the "best" alternative to the [3H] thymidine LPT.
Collapse
Affiliation(s)
- Nirosha Ganesan
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, 3000 Leuven, Belgium
| | - Steven Ronsmans
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium
- Clinic for Occupational and Environmental Medicine, Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Peter Hoet
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, 3000 Leuven, Belgium
- Correspondence:
| |
Collapse
|
6
|
Dadyar M, Hussen BM, Eslami S, Taheri M, Emadi F, Ghafouri-Fard S, Sayad A. Expression of T cell-related lncRNAs in multiple sclerosis. Front Genet 2022; 13:967157. [PMID: 36092928 PMCID: PMC9461285 DOI: 10.3389/fgene.2022.967157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been demonstrated to in the pathophysiology of multiple sclerosis (MS). In order to appraise the role of T cell-related lncRNAs in this disorder, we assessed expressions of NEST, RMRP, TH2-LCR, MAFTRR and FLICR in MS patients and healthy individuals. We detected significant difference in the expression of RMRP and FLICR between cases and controls. There were substantial correlations between expressions of NEST, RMRP, TH2-LCR, MAFTRR and FLICR lncRNAs among patients, but not controls. The strongest correlations were found between RMRP and TH2-LCR, and between MAFTRR and RMRP with correlation coefficients of 0.69 and 0.59, respectively. ROC curve analysis revealed appropriate power of FLICR in differentiating between MS patients and healthy controls (AUC value = 0.84). Expression of NEST lncRNA was positively correlated with disease duration in MS patients, but negatively correlated with age at onset. In brief, we reported dysregulation of two T cell-related lncRNAs in MS patients and proposed FLICR as a putative marker for this disorder.
Collapse
Affiliation(s)
- Maryam Dadyar
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Solat Eslami
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Farhad Emadi
- Skull Base Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Soudeh Ghafouri-Fard, ; Arezou Sayad,
| | - Arezou Sayad
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Soudeh Ghafouri-Fard, ; Arezou Sayad,
| |
Collapse
|
7
|
Docampo MJ, Lutterotti A, Sospedra M, Martin R. Mechanistic and Biomarker Studies to Demonstrate Immune Tolerance in Multiple Sclerosis. Front Immunol 2022; 12:787498. [PMID: 35069562 PMCID: PMC8766750 DOI: 10.3389/fimmu.2021.787498] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
The induction of specific immunological tolerance represents an important therapeutic goal for multiple sclerosis and other autoimmune diseases. Sound knowledge of the target antigens, the underlying pathomechanisms of the disease and the presumed mechanisms of action of the respective tolerance-inducing approach are essential for successful translation. Furthermore, suitable tools and assays to evaluate the induction of immune tolerance are key aspects for the development of such treatments. However, investigation of the mechanisms of action underlying tolerance induction poses several challenges. The optimization of sensitive, robust methods which allow the assessment of low frequency autoreactive T cells and the long-term reduction or change of their responses, the detection of regulatory cell populations and their immune mediators, as well as the validation of specific biomarkers indicating reduction of inflammation and damage, are needed to develop tolerance-inducing approaches successfully to patients. This short review focuses on how to demonstrate mechanistic proof-of-concept in antigen-specific tolerance-inducing therapies in MS.
Collapse
Affiliation(s)
| | | | | | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Neurology Clinic, University Hospital Zurich & University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Ou K, Hamo D, Schulze A, Roemhild A, Kaiser D, Gasparoni G, Salhab A, Zarrinrad G, Amini L, Schlickeiser S, Streitz M, Walter J, Volk HD, Schmueck-Henneresse M, Reinke P, Polansky JK. Strong Expansion of Human Regulatory T Cells for Adoptive Cell Therapy Results in Epigenetic Changes Which May Impact Their Survival and Function. Front Cell Dev Biol 2021; 9:751590. [PMID: 34869339 PMCID: PMC8639223 DOI: 10.3389/fcell.2021.751590] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/12/2021] [Indexed: 12/27/2022] Open
Abstract
Adoptive transfer of regulatory T cells (Treg) is a promising new therapeutic option to treat detrimental inflammatory conditions after transplantation and during autoimmune disease. To reach sufficient cell yield for treatment, ex vivo isolated autologous or allogenic Tregs need to be expanded extensively in vitro during manufacturing of the Treg product. However, repetitive cycles of restimulation and prolonged culture have been shown to impact T cell phenotypes, functionality and fitness. It is therefore critical to scrutinize the molecular changes which occur during T cell product generation, and reexamine current manufacturing practices. We performed genome-wide DNA methylation profiling of cells throughout the manufacturing process of a polyclonal Treg product that has proven safety and hints of therapeutic efficacy in kidney transplant patients. We found progressive DNA methylation changes over the duration of culture, which were donor-independent and reproducible between manufacturing runs. Differentially methylated regions (DMRs) in the final products were significantly enriched at promoters and enhancers of genes implicated in T cell activation. Additionally, significant hypomethylation did also occur in promoters of genes implicated in functional exhaustion in conventional T cells, some of which, however, have been reported to strengthen immunosuppressive effector function in Tregs. At the same time, a set of reported Treg-specific demethylated regions increased methylation levels with culture, indicating a possible destabilization of Treg identity during manufacturing, which was independent of the purity of the starting material. Together, our results indicate that the repetitive TCR-mediated stimulation lead to epigenetic changes that might impact functionality of Treg products in multiple ways, by possibly shifting to an effector Treg phenotype with enhanced functional activity or by risking destabilization of Treg identity and impaired TCR activation. Our analyses also illustrate the value of epigenetic profiling for the evaluation of T cell product manufacturing pipelines, which might open new avenues for the improvement of current adoptive Treg therapies with relevance for conventional effector T cell products.
Collapse
Affiliation(s)
- Kristy Ou
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dania Hamo
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anne Schulze
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andy Roemhild
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel Kaiser
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gilles Gasparoni
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Abdulrahman Salhab
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Ghazaleh Zarrinrad
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Leila Amini
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mathias Streitz
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jörn Walter
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Schmueck-Henneresse
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Reinke
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia K Polansky
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, Berlin, Germany
| |
Collapse
|
9
|
Multiple sclerosis patients have reduced resting and increased activated CD4 +CD25 +FOXP3 +T regulatory cells. Sci Rep 2021; 11:10476. [PMID: 34006899 PMCID: PMC8131694 DOI: 10.1038/s41598-021-88448-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/01/2021] [Indexed: 12/26/2022] Open
Abstract
Resting and activated subpopulations of CD4+CD25+CD127loT regulatory cells (Treg) and CD4+CD25+CD127+ effector T cells in MS patients and in healthy individuals were compared. Peripheral blood mononuclear cells isolated using Ficoll Hypaque were stained with monoclonal antibodies and analysed by flow cytometer. CD45RA and Foxp3 expression within CD4+ cells and in CD4+CD25+CD127loT cells identified Population I; CD45RA+Foxp3+, Population II; CD45RA−Foxp3hi and Population III; CD45RA−Foxp3+ cells. Effector CD4+CD127+ T cells were subdivided into Population IV; memory /effector CD45RA− CD25−Foxp3− and Population V; effector naïve CD45RA+CD25−Foxp3−CCR7+ and terminally differentiated RA+ (TEMRA) effector memory cells. Chemokine receptor staining identified CXCR3+Th1-like Treg, CCR6+Th17-like Treg and CCR7+ resting Treg. Resting Treg (Population I) were reduced in MS patients, both in untreated and treated MS compared to healthy donors. Activated/memory Treg (Population II) were significantly increased in MS patients compared to healthy donors. Activated effector CD4+ (Population IV) were increased and the naïve/ TEMRA CD4+ (Population V) were decreased in MS compared to HD. Expression of CCR7 was mainly in Population I, whereas expression of CCR6 and CXCR3 was greatest in Populations II and intermediate in Population III. In MS, CCR6+Treg were lower in Population III. This study found MS is associated with significant shifts in CD4+T cells subpopulations. MS patients had lower resting CD4+CD25+CD45RA+CCR7+ Treg than healthy donors while activated CD4+CD25hiCD45RA−Foxp3hiTreg were increased in MS patients even before treatment. Some MS patients had reduced CCR6+Th17-like Treg, which may contribute to the activity of MS.
Collapse
|
10
|
Regulatory T Cells for the Induction of Transplantation Tolerance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33523454 DOI: 10.1007/978-981-15-6407-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Organ transplantation is the optimal treatment for terminal and irreversible organ failure. Achieving transplantation tolerance has long been the ultimate goal in the field of transplantation. Regulatory T cell (Treg)-based therapy is a promising novel approach for inducing donor organ-specific tolerance. Tregs play critical roles in the maintenance of immune homeostasis and self-tolerance, by promoting transplantation tolerance through a variety of mechanisms on different target cells, including anti-inflammatory cytokine production, induction of apoptosis, disruption of metabolic pathways, and mutual interaction with dendritic cells. The continued success of Treg-based therapy in the clinical setting is critically dependent on preclinical studies that support its translational potential. However, although some initial clinical trials of adoptive Treg therapy have successively demonstrated safety and efficacy for immunosuppressant minimization and transplantation tolerance induction, most Treg-based hematopoietic stem cell and solid organ clinical trials are still in their infancy. These clinical trials have not only focused on safety and efficacy but also included optimization and standardization protocols of good manufacturing practice regarding cell isolation, expansion, dosing, timing, specificity, quality control, concomitant immunosuppressants, and post-administration monitoring. We herein report a brief introduction of Tregs, including their phenotypic and functional characterization, and focus on the clinical translation of Treg-based therapeutic applications in the setting of transplantation.
Collapse
|
11
|
Cui C, Fan J, Zeng Q, Cai J, Chen Y, Chen Z, Wang W, Li SY, Cui Q, Yang J, Tang C, Xu G, Cai J, Geng B. CD4 + T-Cell Endogenous Cystathionine γ Lyase-Hydrogen Sulfide Attenuates Hypertension by Sulfhydrating Liver Kinase B1 to Promote T Regulatory Cell Differentiation and Proliferation. Circulation 2020; 142:1752-1769. [PMID: 32900241 DOI: 10.1161/circulationaha.119.045344] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Hydrogen sulfide (H2S) has antihypertension and anti-inflammatory effects, and its endogenous-generation key enzyme cystathionine γ lyase (CSE) is expressed in CD4+ T cells. However, the role of CD4+ T-cell endogenous CSE/H2S in the development of hypertension is unclear. METHODS Peripheral blood lymphocytes were isolated from hypertensive patients or spontaneously hypertensive rats, then H2S production and expression of its generation enzymes, cystathionine β synthase and CSE, were measured to determine the major H2S generation system changes in hypertension. Mice with CSE-specific knockout in T cells (conditional knockout, by CD4cre mice hybridization) and CD4 null mice were generated for investigating the pathophysiological relevance of the CSE/H2S system. RESULTS In lymphocytes, H2S from CSE, but not cystathionine β synthase, responded to blood pressure changes, supported by lymphocyte CSE protein changes and a negative correlation between H2S production with systolic blood pressure and diastolic blood pressure, but positive correlation with the serum level of interleukin 10 (an anti-inflammatory cytokine). Deletion of CSE in T cells elevated BP (5-8 mm Hg) under the physiological condition and exacerbated angiotensin II-induced hypertension. In keeping with hypertension, mesenteric artery dilation impaired association with arterial inflammation, an effect attributed to reduced immunoinhibitory T regulatory cell (Treg) numbers in the blood and kidney, thus causing excess CD4+ and CD8+ T cell infiltration in perivascular adipose tissues and kidney. CSE knockout CD4+ T cell transfer into CD4 null mice, also showed the similar phenotypes' confirming the role of endogenous CSE/H2S action. Adoptive transfer of Tregs (to conditional knockout mice) reversed hypertension, vascular relaxation impairment, and immunocyte infiltration, which confirmed that conditional knockout-induced hypertension was attributable, in part, to the reduced Treg numbers. Mechanistically, endogenous CSE/H2S promoted Treg differentiation and proliferation by activating AMP-activated protein kinase. In part, it depended on activation of its upstream kinase, liver kinase B1, by sulfhydration to facilitate its substrate binding and phosphorylation. CONCLUSION The constitutive sulfhydration of liver kinase B1 by CSE-derived H2S activates its target kinase, AMP-activated protein kinase, and promotes Treg differentiation and proliferation, which attenuates the vascular and renal immune-inflammation, thereby preventing hypertension.
Collapse
Affiliation(s)
- Changting Cui
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, P.R. China (C.C., Z.C., W.W., S.y.L., Jun Cai, B.G.)
| | - Jinghui Fan
- Department of Physiology and Pathophysiology, School of Basic Medical Science; Peking University Health Science Center, Beijing, P.R. China (J.F., Q.C., J.Y., C.T., G.X.)
| | - Qiang Zeng
- Health Management Institute, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, P.R. China (Q.Z., Y.C.)
| | - Junyan Cai
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu. P.R. China (Junyan Cai)
| | - Yongzeng Chen
- Health Management Institute, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, P.R. China (Q.Z., Y.C.)
| | - Zhenzhen Chen
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, P.R. China (C.C., Z.C., W.W., S.y.L., Jun Cai, B.G.)
| | - Wenjie Wang
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, P.R. China (C.C., Z.C., W.W., S.y.L., Jun Cai, B.G.)
| | - Shuang Yue Li
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, P.R. China (C.C., Z.C., W.W., S.y.L., Jun Cai, B.G.)
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Science; Peking University Health Science Center, Beijing, P.R. China (J.F., Q.C., J.Y., C.T., G.X.)
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Science; Peking University Health Science Center, Beijing, P.R. China (J.F., Q.C., J.Y., C.T., G.X.)
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Science; Peking University Health Science Center, Beijing, P.R. China (J.F., Q.C., J.Y., C.T., G.X.)
| | - Guoheng Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Science; Peking University Health Science Center, Beijing, P.R. China (J.F., Q.C., J.Y., C.T., G.X.)
| | - Jun Cai
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, P.R. China (C.C., Z.C., W.W., S.y.L., Jun Cai, B.G.)
| | - Bin Geng
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, P.R. China (C.C., Z.C., W.W., S.y.L., Jun Cai, B.G.)
| |
Collapse
|
12
|
Bao Z, Li J, Zhang P, Pan Q, Liu B, Zhu J, Jian Q, Jia D, Yi C, Moeller CJ, Liu H. Toll-Like Receptor 3 Activator Preconditioning Enhances Modulatory Function of Adipose‑Derived Mesenchymal Stem Cells in a Fully MHC-Mismatched Murine Model of Heterotopic Heart Transplantation. Ann Transplant 2020; 25:e921287. [PMID: 32366814 PMCID: PMC7219555 DOI: 10.12659/aot.921287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Donor-specific tolerance is the ultimate goal in organ transplantation. Diverse approaches, including the use of mesenchymal stem cells (MSCs), have been investigated to induce graft tolerance. Non-stimulated MSCs showed limited regulatory functions through interaction with multiple immune-regulatory cells, such as regulatory T cells (Tregs). To augment their functions, MSCs have been preconditioned with toll-like receptor (TLR3/4) agonist in autoimmune disease models, but results were conflicting. Material/Methods We evaluated the immunomodulatory effects of mouse adipose-derived mesenchymal stem cells (ADSCs) preconditioned with various combinations of TLR3/4 agonist and antagonists, including polyinosinic-polycytidylic acid poly(I:C)-TLR3 agonist, lipopolysaccharide (LPS) -TLR4 agonist, and TAK242-TLR4 antagonist. In vitro and in vivo experiments including mixed lymphocyte reaction, cytokines measurement, Tregs analysis, and a fully mismatched MHC heterotopic heart transplantation in mice (BALB/c to C57BL/6) were conducted. Results ADSCs preconditioned with poly(I:C) showed the highest efficiency in inhibiting lymphocyte proliferation, which was correlated with the upregulation of fibrinogen-like protein 2 (FGL2), an effector molecule of Tregs. The mean survival of cardiac allografts was extended from 8 to 12 days by intravenous injection of a single dose of ADSCs preconditioned with TLR3 agonist. The proportion of Tregs in the recipient’s spleen was significantly increased by injecting the poly(I:C)-stimulated ADSCs. Conclusions These results show that short-term TLR3 agonist preconditioning enhances the immunomodulatory efficacy of ADSCs, which can induce the generation of Tregs and upregulate the expression of FGL2, thereby improving the outcome of patients receiving organ transplantation.
Collapse
Affiliation(s)
- Zhiye Bao
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China (mainland)
| | - Jingjing Li
- Department of Pediatric Surgery, Tianjin Children's Hospital, Tianjin, China (mainland)
| | - Pengju Zhang
- Oncology Center of People's Liberation Army (PLA), 81st Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Qi Pan
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China (mainland)
| | - Boqian Liu
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China (mainland)
| | - Jiayi Zhu
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China (mainland)
| | - Qian Jian
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China (mainland)
| | - Degong Jia
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China (mainland)
| | - Caiyu Yi
- China Medical University, Shenyang, Liaoning, China (mainland)
| | | | - Hao Liu
- The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
13
|
Beauford SS, Kumari A, Garnett-Benson C. Ionizing radiation modulates the phenotype and function of human CD4+ induced regulatory T cells. BMC Immunol 2020; 21:18. [PMID: 32299365 PMCID: PMC7164225 DOI: 10.1186/s12865-020-00349-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The use of immunotherapy strategies for the treatment of advanced cancer is rapidly increasing. Most immunotherapies rely on induction of CD8+ tumor-specific cytotoxic T cells that are capable of directly killing cancer cells. Tumors, however, utilize a variety of mechanisms that can suppress anti-tumor immunity. CD4+ regulatory T cells can directly inhibit cytotoxic T cell activity and these cells can be recruited, or induced, by cancer cells allowing escape from immune attack. The use of ionizing radiation as a treatment for cancer has been shown to enhance anti-tumor immunity by several mechanisms including immunogenic tumor cell death and phenotypic modulation of tumor cells. Less is known about the impact of radiation directly on suppressive regulatory T cells. In this study we investigate the direct effect of radiation on human TREG viability, phenotype, and suppressive activity. RESULTS Both natural and TGF-β1-induced CD4+ TREG cells exhibited increased resistance to radiation (10 Gy) as compared to CD4+ conventional T cells. Treatment, however, decreased Foxp3 expression in natural and induced TREG cells and the reduction was more robust in induced TREGS. Radiation also modulated the expression of signature iTREG molecules, inducing increased expression of LAG-3 and decreased expression of CD25 and CTLA-4. Despite the disconcordant modulation of suppressive molecules, irradiated iTREGS exhibited a reduced capacity to suppress the proliferation of CD8+ T cells. CONCLUSIONS Our findings demonstrate that while human TREG cells are more resistant to radiation-induced death, treatment causes downregulation of Foxp3 expression, as well as modulation in the expression of TREG signature molecules associated with suppressive activity. Functionally, irradiated TGF-β1-induced TREGS were less effective at inhibiting CD8+ T cell proliferation. These data suggest that doses of radiotherapy in the hypofractionated range could be utilized to effectively target and reduce TREG activity, particularly when used in combination with cancer immunotherapies.
Collapse
Affiliation(s)
- Samantha S Beauford
- Department of Biology, Georgia State University, 161 Jesse Hill Jr. Dr, Atlanta, GA, 30303, USA
| | - Anita Kumari
- Department of Biology, Georgia State University, 161 Jesse Hill Jr. Dr, Atlanta, GA, 30303, USA
| | - Charlie Garnett-Benson
- Department of Biology, Georgia State University, 161 Jesse Hill Jr. Dr, Atlanta, GA, 30303, USA.
| |
Collapse
|
14
|
|
15
|
Gowayed MA, Rothe K, Rossol M, Attia AS, Wagner U, Baerwald C, El-Abhar HS, Refaat R. The role of α7nAChR in controlling the anti-inflammatory/anti-arthritic action of galantamine. Biochem Pharmacol 2019; 170:113665. [PMID: 31606410 DOI: 10.1016/j.bcp.2019.113665] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/08/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The evolution of the "cholinergic anti-inflammatory pathway" and the fact that the α 7 subunit of the nicotinic acetylcholine receptor (α7nAChR) is present in the spleen, joint and on the surface of lymphocytes, opened up the prospective in this study of targeting the α7nAChR by the anticholinesterase and cholinergic drug, galantamine, to control inflammation in rheumatoid arthritis (RA). METHODS Twelve-adjuvant arthritic rats were exposed to the selective α7nAChR blocker methylcaconitine citrate 15 min before galantamine treatment. As control, six adjuvant arthritic rats were treated with galantamine and six others were untreated. After five days TNF-α levels were assessed in spleen and joints, while reduced glutathione was measured in blood and joint tissue. In the second part, magnetically sorted CD4 + T cells from peripheral blood mononuclear cells of RA patients and healthy donors were used to sort CD4 + CD25 - primary T cells (Tresp) and CD4 + CD25 + CD127low Tregs. The suppressive function of Tregs was investigated after incubation with galantamine using flow cytometry. Cell culture supernatants were analyzed for TNF-α and IL-10 levels after three days incubation period of Tregs with Tresp. The effect of galantamine on Tregs was then blocked by α-Bungarotoxin and the same assay has been repeated. RESULTS & CONCLUSION Selective α7nAChR blockade interrupted the anti-inflammatory effect of galantamine in the spleen and joints of arthritic rats. In healthy donors, galantamine could strengthen the suppressive activity of Tregs; while in RA patients it did not modulate the function of Tregs significantly. Further studies are necessary to investigate whether modulation of the cholinergic nervous system, especially α7nAChR, could have impact on the disturbed immune system in RA, which may open up a new treatment option of autoimmune diseases.
Collapse
Affiliation(s)
- Mennatallah A Gowayed
- Lecturer of Pharmacology, Department of Pharmacology and Therapeutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Egypt.
| | - Kathrin Rothe
- Department of Internal Medicine, Division of Rheumatology, University of Leipzig, Germany
| | - Manuela Rossol
- Department of Internal Medicine, Division of Rheumatology, University of Leipzig, Germany
| | - Ahmed S Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Egypt
| | - Ulf Wagner
- Department of Internal Medicine, Division of Rheumatology, University Hospital Leipzig, Germany
| | - Christoph Baerwald
- Department of Internal Medicine, Division of Rheumatology, University Hospital Leipzig, Germany
| | - Hanan S El-Abhar
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Egypt
| | - Rowaida Refaat
- Department of Pharmacology, Medical Research Institute, University of Alexandria, Egypt
| |
Collapse
|
16
|
Ma F, Li S, Gao X, Zhou J, Zhu X, Wang D, Cai Y, Li F, Yang Q, Gu X, Ge W, Liu H, Xiao X, Hao H. Interleukin-6-mediated CCR9 + interleukin-17-producing regulatory T cells polarization increases the severity of necrotizing enterocolitis. EBioMedicine 2019; 44:71-85. [PMID: 31129099 PMCID: PMC6604880 DOI: 10.1016/j.ebiom.2019.05.042] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 01/03/2023] Open
Abstract
Background Increased frequency of CCR9+ CD4+ T cells in peripheral blood is linked to several gastrointestinal inflammatory diseases; however, its relationship with necrotizing enterocolitis (NEC) is not understood. We investigated whether the frequencies of CCR9+ CD4+ T cells and related subsets were increased in peripheral blood of both patients and mice with NEC. Methods CCR9+ CD4+ T cells and related subsets were evaluated by flow cytometry in peripheral blood collected from both patients and mice with NEC and controls. The suppressive function of CCR9+ regulatory T (Treg) cells in NEC was assessed via in vitro suppression assay. An in vitro T cell polarization assay was performed to investigate the role of proinflammatory cytokines in Treg cell polarization. In vivo Treg cell polarization analysis was performed using NEC mice treated with anti-interleukin-6 (IL-6) receptor antibody. Findings A higher proportion of CCR9+ CD4+ T cells occurred in peripheral blood of both patients and mice with NEC than in controls. Elevated CCR9+ CD4+ T cells were primarily CCR9+ IL-17-producing Treg cells, possessing features of conventional Treg cells, but their suppressive activity was seriously impaired and negatively correlated with the severity of intestinal tissue injury. IL-6 promoted polarization of CCR9+ Treg cells to CCR9+ IL-17-producing Treg cells, and blocking IL-6 signalling inhibited this conversion in vitro and ameliorated experimental NEC in vivo. Interpretation Collectively, these data suggested that CCR9+ IL-17-producing Treg cells may be a biomarker of severity and highlight the possibility that antibodies targeting IL-6R could ameliorate NEC by modulating lymphocyte balance. Fund This work was supported by the Science and Technology Planning Project of Guangdong Province, China (2017A020215100), the Science and Technology Foundation of Guangzhou, China (201704020086 and 201604020154), the Medical Scientific Research Foundation of Guangdong Province, China (A2017304 and A2014704), and the Social Science and Technology Development Foundation of Dongguan, China (2016108101037).
Collapse
Affiliation(s)
- Fei Ma
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sitao Li
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Gao
- Department of Neonatology, the Foshan Women and Children Hospital, Foshan, China
| | - Jialiang Zhou
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xiaochun Zhu
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Desheng Wang
- Department of Neonatology, the Fifth People's Hospital of Dongguan, Dongguan, China
| | - Yao Cai
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fei Li
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiuping Yang
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xia Gu
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wuping Ge
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Huanliang Liu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Xin Xiao
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Hu Hao
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
17
|
Wendering DJ, Amini L, Schlickeiser S, Reinke P, Volk HD, Schmueck-Henneresse M. The Value of a Rapid Test of Human Regulatory T Cell Function Needs to be Revised. Front Immunol 2019; 10:150. [PMID: 30804944 PMCID: PMC6370705 DOI: 10.3389/fimmu.2019.00150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/17/2019] [Indexed: 11/15/2022] Open
Abstract
CD4+CD25+FoxP3+ human regulatory TCELLS (TREG) are promising candidates for reshaping undesired immunity/inflammation by adoptive cell transfer, yet their application is strongly dependent on robust assays testing their functionality. Several studies along with first clinical data indicate TREG to be auspicious to use for future cell therapies, e.g., to induce tolerance after solid organ transplantation. To this end, TREG suppressive capacity has to be thoroughly evaluated prior to any therapeutic application. A 7 h-protocol for the assessment of TREG function by suppression of the early activation markers CD154 and CD69 on CD4+CD25− responder TCELLS (TRESP) upon polyclonal stimulation via αCD3/28-coated activating microbeads has previously been published. Even though this assay has since been applied by various groups, the protocol comes with a critical pitfall, which is yet not corrected by the journal of its original publication. Our results demonstrate that the observed decrease in activation marker frequency on TRESP is due to competition for αCD3/28-coated microbeads as opposed to a TREG-attributable effect and therefore the protocol cannot further be used as a diagnostic test to assess suppressive TREG function.
Collapse
Affiliation(s)
- Desiree J Wendering
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Centrum for Regenerative Therapies (B-CRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Leila Amini
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Centrum for Regenerative Therapies (B-CRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stephan Schlickeiser
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Centrum for Regenerative Therapies (B-CRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Reinke
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Centrum for Regenerative Therapies (B-CRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Dieter Volk
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Centrum for Regenerative Therapies (B-CRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Schmueck-Henneresse
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Centrum for Regenerative Therapies (B-CRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
18
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J Cell Mol Med 2018; 23:720-730. [PMID: 30484934 PMCID: PMC6349184 DOI: 10.1111/jcmm.14035] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/20/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) is a common, severe emergency case in clinics, with high incidence, significant mortality and increased costs. Despite development in the understanding of its pathophysiology, the therapeutic choices are still confined to dialysis and renal transplantation. Considering their antiapoptotic, immunomodulatory, antioxidative and pro‐angiogenic effects, mesenchymal stem cells (MSCs) may be a promising candidate for AKI management. Based on these findings, some clinical trials have been performed, but the results are contradictory (NCT00733876, NCT01602328). The low engraftment, poor survival rate, impaired paracrine ability and delayed administration of MSCs are the four main reasons for the limited clinical efficacy. Investigators have developed a series of preconditioning strategies to improve MSC survival rates and paracrine ability. In this review, by summarizing these encouraging studies, we intend to provide a comprehensive understanding of various preconditioning strategies on AKI therapy and improve the prognosis of AKI patients by regenerative medicine.
Collapse
Affiliation(s)
- Lingfei Zhao
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ping Zhang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Hua Jiang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jianghua Chen
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
19
|
Achita P, Dervovic D, Ly D, Lee JB, Haug T, Joe B, Hirano N, Zhang L. Infusion of ex-vivo expanded human TCR-αβ + double-negative regulatory T cells delays onset of xenogeneic graft-versus-host disease. Clin Exp Immunol 2018; 193:386-399. [PMID: 30066399 DOI: 10.1111/cei.13145] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2018] [Indexed: 12/25/2022] Open
Abstract
Despite the demonstration of potent immunosuppressive function of T cell receptor (TCR)-αβ+ double-negative regulatory T cells (DN Tregs ), scarce numbers and lack of effective expansion method limit their clinical applications. Here we describe an approach that allows for ∼3500-fold ex-vivo expansion of human DN Tregs within 3 weeks with > 97% purity. Ex-vivo-expanded DN Tregs suppress proliferation of polyclonally stimulated autologous T and B cells in vitro through direct cell-to-cell contact. In vivo, we demonstrate for the first time that infusion of human DN Tregs delayed an onset of xenogeneic graft-versus-host disease (GVHD) significantly in a humanized mouse model. Furthermore, preincubation of ex-vivo-expanded DN Tregs with a mechanistic target of rapamycin (mTOR) inhibitor rapamycin enhanced their immune regulatory function further. Taken together, this study demonstrates that human DN Tregs can be expanded ex vivo to therapeutic numbers. The expanded DN Tregs can suppress proliferation of T and B cells and attenuate GVHD, highlighting the potential clinical use of DN Tregs to mitigate GVHD.
Collapse
Affiliation(s)
- P Achita
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, Toronto, ON, Canada
| | - D Dervovic
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - D Ly
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - J B Lee
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - T Haug
- Department of Internal Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - B Joe
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - N Hirano
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - L Zhang
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Jiang H, Xin S, Yan Y, Lun Y, Yang X, Zhang J. Abnormal acetylation of FOXP3 regulated by SIRT-1 induces Treg functional deficiency in patients with abdominal aortic aneurysms. Atherosclerosis 2018. [DOI: 10.1016/j.atherosclerosis.2018.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
21
|
Chenna Narendra S, Chalise JP, Biggs S, Kalinke U, Magnusson M. Regulatory T-Cells Mediate IFN-α-Induced Resistance against Antigen-Induced Arthritis. Front Immunol 2018. [PMID: 29515584 PMCID: PMC5826073 DOI: 10.3389/fimmu.2018.00285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective CD4+FoxP3+CD25+ regulatory T-cells (Tregs) are important for preventing tissue destruction. Here, we investigate the role of Tregs for protection against experimental arthritis by IFN-α. Methods Arthritis was triggered by intra-articular injection of methylated bovine serum albumin (mBSA) in wild-type mice, Foxp3DTReGFP+/− mice [allowing selective depletion of Tregs by diphtheria toxin (DT)] and CD4-Cre+/− IFNA1R flox/flox mice (devoid of IFNAR signaling in T-cells) earlier immunized with mBSA, with or without treatment with IFN-α or the indoleamine 2,3-dioxygenase (IDO)-metabolite kynurenine. Tregs were depleted in DT-treated Foxp3DTReGFP+/− mice and enumerated by FoxP3 staining. Suppressive capacity of FACS-sorted CD25+highCD4+ Tregs was tested in vivo by adoptive transfer and ex vivo in cocultures with antigen-stimulated CFSE-stained T-responder (CD25−CD4+) cells. IDO was inhibited by 1-methyl tryptophan. Results Both control mice and mice devoid of IFNAR-signaling in T helper cells were protected from arthritis by IFN-α. Depletion of Tregs in the arthritis phase, but not at immunization, abolished the protective effect of IFN-α and kynurenine against arthritis. IFN-α increased the number of Tregs in ex vivo cultures upon antigen recall stimulation but not in naïve cells. IFN-α also increased the suppressive capacity of Tregs against mBSA-induced T-responder cell proliferation ex vivo and against arthritis when adoptively transferred. The increased suppressive activity against proliferation conferred by IFN-α was clearly reduced by in vivo inhibition of IDO at immunization, which also abolished the protective effect of IFN-α against arthritis. Conclusion By activating IDO during antigen sensitization, IFN-α activates Tregs, which prevent arthritis triggered by antigen rechallenge. This is one way by which IFN-α suppresses inflammation.
Collapse
Affiliation(s)
- Sudeep Chenna Narendra
- Division of Rheumatology, Autoimmunity and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | | - Sophie Biggs
- Division of Rheumatology, Autoimmunity and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Ulrich Kalinke
- Twincore, Zentrum für Experimentelle und Klinische Infektionsforschung, Hannover, Germany
| | - Mattias Magnusson
- Division of Rheumatology, Autoimmunity and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
22
|
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
|
23
|
Yang X, Lun Y, Jiang H, Liu X, Duan Z, Xin S, Zhang J. SIRT1-Regulated Abnormal Acetylation of FOXP3 Induces Regulatory T-Cell Function Defect in Hashimoto's Thyroiditis. Thyroid 2018; 28:246-256. [PMID: 29336238 DOI: 10.1089/thy.2017.0286] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Hashimoto's thyroiditis (HT) is an autoimmune thyroid disease characterized by low expression of transcription factor Forkhead Box P3 (FOXP3) and functional deficiency of a cluster of differentiation regulatory T cells (Tregs). This study aimed to investigate the mechanism of Treg dysfunction in HT. METHODS The number of CD4+CD25+FOXP3+ T cells was determined by flow cytometry. Expression of FOXP3 and Sirtuin type 1 (SIRT1) was evaluated by Western blot analysis. Acetylation of FOXP3 was analyzed by immunoprecipitation and Western blot analysis. The suppressive function of Treg was analyzed by the 5,6-carboxyfluorescein diacetate succinimidyl ester (CFSE) assay. RESULTS The percentage of CD4+CD25+FOXP3+ T cells, expression of FOXP3, and FOXP3 acetylation level in the HT group were significantly lower than in the control groups. Conversely, SIRT1 expression was significantly higher in the HT group than in the other two groups. After Ex-527 treatment, the CD4+CD25+FOXP3+ T cells percentage, FOXP3 expression, and FOXP3 acetylation level in the HT group were significantly increased. HT Tregs exhibited less suppressive activity, but Ex-527 treatment significantly increased their suppressive activity. CONCLUSIONS The findings demonstrate that the reduced FOXP3 expression level and Treg function defect in HT patients are regulated by SIRT1-mediated abnormal FOXP3 acetylation. Ex-527 may upregulate the FOXP3 acetylation level and subsequently increase the number and suppressive function of Treg cells.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Vascular Surgery, The First Hospital of China Medical University , Shenyang, P.R. China
| | - Yu Lun
- Department of Vascular Surgery, The First Hospital of China Medical University , Shenyang, P.R. China
| | - Han Jiang
- Department of Vascular Surgery, The First Hospital of China Medical University , Shenyang, P.R. China
| | - Xun Liu
- Department of Vascular Surgery, The First Hospital of China Medical University , Shenyang, P.R. China
| | - Zhiquan Duan
- Department of Vascular Surgery, The First Hospital of China Medical University , Shenyang, P.R. China
| | - Shijie Xin
- Department of Vascular Surgery, The First Hospital of China Medical University , Shenyang, P.R. China
| | - Jian Zhang
- Department of Vascular Surgery, The First Hospital of China Medical University , Shenyang, P.R. China
| |
Collapse
|
24
|
Yao Y, Li H, Chen J, Xu W, Yang G, Bao Z, Xia D, Lu G, Hu S, Zhou J. TREM-2 serves as a negative immune regulator through Syk pathway in an IL-10 dependent manner in lung cancer. Oncotarget 2018; 7:29620-34. [PMID: 27102437 PMCID: PMC5045421 DOI: 10.18632/oncotarget.8813] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/28/2016] [Indexed: 01/31/2023] Open
Abstract
During infection, triggering receptor expressed on myeloid cells-2 (TREM-2) restrains dendritic cells (DCs) and macrophages (MΦs) phagocytosis, as well as reduces pro-inflammatory cytokines release through DNAX-activation protein 12 (DAP12) signaling. However, the role of TREM-2 signaling in cancer has never been elucidated. In the current study, we found that TREM-2 was up-regulated on peripheral blood monocytes in tumor-bearing host. More TREM-2+DCs were detected in the lung of 3LL tumor-bearing mice. On the other hand, the level of TREM-2 on pulmonary MΦs positively correlated with the pathological staging of lung cancer. However, surgical or chemotherapeutic reduction of tumor burden led to the obvious decline of TREM-2. In vitro, TREM-2 expression of bone marrow (BM)-derived DCs and MΦs was induced by conditional medium (CM) containing the supernatant of 3LL cells. TREM-2+DCs from CM and/or tumor-bearing mice held altered phenotypes (CD80LowCD86LowMHCIILow) and impaired functions, such as, reduced interleukin (IL)-12 secretion, increased IL-10 production, and weakened ovalbumin (OVA)-endocytic capacity; also developed potent inhibitory effect on T cell proliferation that could be partially reversed by TREM-2 blockage. Moreover, spleen tyrosine kinase (Syk) inhibitor restrained IL-10 production of TREM-2+DC. Remarkably, IL-10 neutralizing antibody and Syk inhibitor both lowered the suppressive potential of TREM-2+DCs in T cell proliferation. Also, adoptive transfer of this TREM-2+DCs accelerated the tumor growth rather than jeopardized survival in lung cancer-bearing mice. In conclusion, these results indicate that TREM-2 might act as a negative immuno-regulatory molecule through Syk pathway in an IL-10 dependent manner and partially predicts prognosis in lung cancer patients.
Collapse
Affiliation(s)
- Yinan Yao
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Hequan Li
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Junjun Chen
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Weiyi Xu
- Department of Clinical Laboratory, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Guangdie Yang
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Zhang Bao
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Dajing Xia
- School of Public Health, Zhejiang University, Institute of Immunology, Zhejiang University, Hangzhou, China.,Institute of Immunology, Zhejiang University, Hangzhou, China
| | - Guohua Lu
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Shuwen Hu
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Ten Brinke A, Marek-Trzonkowska N, Mansilla MJ, Turksma AW, Piekarska K, Iwaszkiewicz-Grześ D, Passerini L, Locafaro G, Puñet-Ortiz J, van Ham SM, Hernandez-Fuentes MP, Martínez-Cáceres EM, Gregori S. Monitoring T-Cell Responses in Translational Studies: Optimization of Dye-Based Proliferation Assay for Evaluation of Antigen-Specific Responses. Front Immunol 2017; 8:1870. [PMID: 29312346 PMCID: PMC5742609 DOI: 10.3389/fimmu.2017.01870] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/08/2017] [Indexed: 01/05/2023] Open
Abstract
Adoptive therapy with regulatory T cells or tolerance-inducing antigen (Ag)-presenting cells is innovative and promising therapeutic approach to control undesired and harmful activation of the immune system, as observed in autoimmune diseases, solid organ and bone marrow transplantation. One of the critical issues to elucidate the mechanisms responsible for success or failure of these therapies and define the specificity of the therapy is the evaluation of the Ag-specific T-cell responses. Several efforts have been made to develop suitable and reproducible assays. Here, we focus on dye-based proliferation assays. We highlight with practical examples the fundamental issues to take into consideration for implementation of an effective and sensitive dye-based proliferation assay to monitor Ag-specific responses in patients. The most critical points were used to design a road map to set up and analyze the optimal assay to assess Ag-specific T-cell responses in patients undergoing different treatments. This is the first step to optimize monitoring of tolerance induction, allowing comparison of outcomes of different clinical studies. The road map can also be applied to other therapeutic interventions, not limited to tolerance induction therapies, in which Ag-specific T-cell responses are relevant such as vaccination approaches and cancer immunotherapy.
Collapse
Affiliation(s)
- Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Natalia Marek-Trzonkowska
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Maria J. Mansilla
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Annelies W. Turksma
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Karolina Piekarska
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Laura Passerini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Grazia Locafaro
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Joan Puñet-Ortiz
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - S. Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | | | - Eva M. Martínez-Cáceres
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
26
|
Zongyi Y, Funian Z, Hao L, Xin W, Ying C, Jialin Z, Yongfeng L, Baifeng L. Interleukin-35 mitigates the function of murine transplanted islet cells via regulation of Treg/Th17 ratio. PLoS One 2017; 12:e0189617. [PMID: 29236782 PMCID: PMC5728515 DOI: 10.1371/journal.pone.0189617] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/29/2017] [Indexed: 12/19/2022] Open
Abstract
Pancreatic islet transplantation is a promising treatment for type 1 diabetes (T1D). Interleukin-35 (IL-35) is a recently discovered cytokine that exhibits potent immunosuppressive functions. However, the role of IL-35 in islet transplant rejection remains to be elucidated. In this study, we isolated islet cells of BALB/c mouse and purified CD4+ T cell subsets of a C57BL/6 mouse. The model for islet transplantation was established in vitro by co-culture of the islet cells and CD4+ T cells. IL-35 (20 ng/ml) was administered every other day. Following co-culture, the islet function and Treg/Th17 ratio were analyzed on days 1, 3, and 5. Furthermore, the Th17/Treg ratio was modulated (1:0–2), and the function of islet cells as well as proliferation of Th17 cells were analyzed. T cell sorting was performed using the magnetic bead sorting method; Treg and Th17 count using flow cytometry; cell proliferation detection using the carboxyfluorescein diacetate succinimidyl ester (CFSE) method, and islet function test using the sugar stimulation test. Results showed that Th17 counts increased in the co-culture system. However, after administration of IL-35, the number of Treg cells increased significantly compared to that in the control group (50.7% of total CD4+ T cells on day 5 in IL-35 group vs. 9.5% in control group) whereas the proliferation rate of Th17 cells was significantly inhibited (0.3% in IL-35 group vs. 7.2% in control group on day 5). Reducing the Th17/Treg ratio significantly improved the function of transplanted islets. Treg inhibited Th17 proliferation and IL-35 enhanced this inhibitory effect. IL-35 mitigates the function of murine transplanted islet cells via regulation of the Treg/Th17 ratio. This might serve as a potential therapeutic strategy for in-vivo islet transplant rejection and T1D.
Collapse
Affiliation(s)
- Yin Zongyi
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen, China
| | - Zou Funian
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Li Hao
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Wang Xin
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Cheng Ying
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China
- Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| | - Zhang Jialin
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China
- Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| | - Liu Yongfeng
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China
- Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| | - Li Baifeng
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China
- Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
- * E-mail:
| |
Collapse
|
27
|
Bai M, Zhang L, Fu B, Bai J, Zhang Y, Cai G, Bai X, Feng Z, Sun S, Chen X. IL-17A improves the efficacy of mesenchymal stem cells in ischemic-reperfusion renal injury by increasing Treg percentages by the COX-2/PGE2 pathway. Kidney Int 2017; 93:814-825. [PMID: 29132705 DOI: 10.1016/j.kint.2017.08.030] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/11/2017] [Accepted: 08/17/2017] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are effective for the management of experimental ischemia-reperfusion acute kidney injury (IRI-AKI). Immune modulation is one of the important mechanisms of MSCs treatment. Interleukin-17A (IL-17A) pretreated MSCs are more immunosuppressive with minimal changes in immunogenicity in vitro. Here, we demonstrated that administration of IL-17A-pretreated MSCs resulted in significantly lower acute tubular necrosis scores, serum creatinine, and BUN of mice with IRI-AKI, compared with the administration of MSCs. Of the co-cultured splenocytes, IL-17A-pretreated MSCs significantly increased the percentages of CD4+Foxp3+ Tregs and decreased concanavalin A-induced T cell proliferation. Furthermore, mice with IRI-AKI that underwent IL-17A-pretreated MSC therapy had significantly lower serum IL-6, TNF-α, and IFN-γ levels, a higher serum IL-10 level, and higher spleen and kidney Treg percentages than the mice that underwent MSCs treatment. Additionally, the depletion of Tregs by PC61 (anti-CD25 antibody) reversed the enhanced treatment efficacy of the IL-17A-pretreatedMSCs on mice with IRI-AKI. Additionally, IL-17A upregulated COX-2 expression and increased PGE2 production. The blockage of COX-2 by celecoxib reversed the benefit of IL-pretreated 17A-MSCs on the serum PGE2 concentration, spleen and kidney Tregs percentages, serum creatinine and BUN levels, renal acute tubular necrosis scores, and serum IL-6, TNF-α, IFN-γ, and IL-10 levels of IRI-pretreated mice with AKI, compared with MSCs. Thus, our results suggest that IL-17A pretreatment enhances the efficacy of MSCs on mice with IRI-AKI by increasing the Treg percentages through the COX-2/PGE2 pathway.
Collapse
Affiliation(s)
- Ming Bai
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China; Department of Nephrology, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Li Zhang
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Bo Fu
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Jiuxu Bai
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Yingjie Zhang
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Guangyan Cai
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Xueyuan Bai
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Zhe Feng
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, the Fourth Military Medical University, Xi'an, China.
| | - Xiangmei Chen
- State Key Laboratory of Kidney Disease, Department of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China.
| |
Collapse
|
28
|
Kim HJ, Cha GS, Joo JY, Lee J, Kim SJ, Lee J, Park SY, Choi J. Targeting the epitope spreader Pep19 by naïve human CD45RA + regulatory T cells dictates a distinct suppressive T cell fate in a novel form of immunotherapy. J Periodontal Implant Sci 2017; 47:292-311. [PMID: 29093987 PMCID: PMC5663667 DOI: 10.5051/jpis.2017.47.5.292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022] Open
Abstract
Purpose Beyond the limited scope of non-specific polyclonal regulatory T cell (Treg)-based immunotherapy, which depends largely on serendipity, the present study explored a target Treg subset appropriate for the delivery of a novel epitope spreader Pep19 antigen as part of a sophisticated form of immunotherapy with defined antigen specificity that induces immune tolerance. Methods Human polyclonal CD4+CD25+CD127lo− Tregs (127-Tregs) and naïve CD4+CD25+CD45RA+ Tregs (45RA-Tregs) were isolated and were stimulated with target peptide 19 (Pep19)-pulsed dendritic cells in a tolerogenic milieu followed by ex vivo expansion. Low-dose interleukin-2 (IL-2) and rapamycin were added to selectively exclude the outgrowth of contaminating effector T cells (Teffs). The following parameters were investigated in the expanded antigen-specific Tregs: the distinct expression of the immunosuppressive Treg marker Foxp3, epigenetic stability (demethylation in the Treg-specific demethylated region), the suppression of Teffs, expression of the homing receptors CD62L/CCR7, and CD95L-mediated apoptosis. The expanded Tregs were adoptively transferred into an NOD/scid/IL-2Rγ−/− mouse model of collagen-induced arthritis. Results Epitope-spreader Pep19 targeting by 45RA-Tregs led to an outstanding in vitro suppressive T cell fate characterized by robust ex vivo expansion, the salient expression of Foxp3, high epigenetic stability, enhanced T cell suppression, modest expression of CD62L/CCR7, and higher resistance to CD95L-mediated apoptosis. After adoptive transfer, the distinct fate of these T cells demonstrated a potent in vivo immunotherapeutic capability, as indicated by the complete elimination of footpad swelling, prolonged survival, minimal histopathological changes, and preferential localization of CD4+CD25+ Tregs at the articular joints in a mechanistic and orchestrated way. Conclusions We propose human naïve CD4+CD25+CD45RA+ Tregs and the epitope spreader Pep19 as cellular and molecular targets for a novel antigen-specific Treg-based vaccination against collagen-induced arthritis.
Collapse
Affiliation(s)
- Hyun-Joo Kim
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Pusan National University School of Dentistry, Yangsan, Korea
| | - Gil Sun Cha
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Pusan National University School of Dentistry, Yangsan, Korea
| | - Ji-Young Joo
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Pusan National University School of Dentistry, Yangsan, Korea
| | - Juyoun Lee
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Pusan National University School of Dentistry, Yangsan, Korea
| | - Sung-Jo Kim
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Pusan National University School of Dentistry, Yangsan, Korea
| | - Jeongae Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Korea
| | - So Youn Park
- Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Korea
| | - Jeomil Choi
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Pusan National University School of Dentistry, Yangsan, Korea
| |
Collapse
|
29
|
Wang C, Zhang N, Qi L, Yuan J, Wang K, Wang K, Ma S, Wang H, Lou W, Hu P, Awais M, Cao S, Fu ZF, Cui M. Myeloid-Derived Suppressor Cells Inhibit T Follicular Helper Cell Immune Response in Japanese Encephalitis Virus Infection. THE JOURNAL OF IMMUNOLOGY 2017; 199:3094-3105. [DOI: 10.4049/jimmunol.1700671] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/17/2017] [Indexed: 12/23/2022]
|
30
|
Sangiorgi B, De Freitas HT, Schiavinato JLDS, Leão V, Haddad R, Orellana MD, Faça VM, Ferreira GA, Covas DT, Zago MA, Panepucci RA. DSP30 enhances the immunosuppressive properties of mesenchymal stromal cells and protects their suppressive potential from lipopolysaccharide effects: A potential role of adenosine. Cytotherapy 2017; 18:846-59. [PMID: 27260206 DOI: 10.1016/j.jcyt.2016.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 04/06/2016] [Accepted: 04/13/2016] [Indexed: 01/09/2023]
Abstract
Multipotent mesenchymal stromal cells (MSC) are imbued with an immunosuppressive phenotype that extends to several immune system cells. In this study, we evaluated how distinct Toll-like receptor (TLR) agonists impact immunosuppressive properties of bone marrow (BM)-MSC and explored the potential mechanisms involved. We show that TLR4 stimulation by lipopolysaccharide (LPS) restricted the ability of MSC to suppress the proliferation of T lymphocytes, increasing the gene expression of interleukin (IL)-1β and IL-6. In contrast, stimulation of TLR9 by DSP30 induced proliferation and the suppressive potential of BM-MSC, coinciding with reducing tumor necrosis factor (TNF)-α expression, increased expression of transforming growth factor (TGF)-β1, increased percentages of BM-MSC double positive for the ectonucleotidases CD39+CD73+ and adenosine levels. Importantly, following simultaneous stimulation with LPS and DSP30, BM-MSC's ability to suppress T lymphocyte proliferation was comparable with that of non-stimulated BM-MSC levels. Moreover, stimulation of BM-MSC with LPS reduced significantly the gene expression levels, on co-cultured T lymphocyte, of IL-10 and interferon (IFN)γ, a cytokine with potential to enhance the immunosuppression mediated by MSC and ameliorate the clinical outcome of patients with graft-versus-host disease (GVHD). Altogether, our findings reiterate the harmful effects of LPS on MSC immunosuppression, besides indicating that DSP30 could provide a protective effect against LPS circulating in the blood of GVHD patients who receive BM-MSC infusions, ensuring a more predictable immunosuppressive effect. The novel effects and potential mechanisms following the stimulation of BM-MSC by DSP30 might impact their clinical use, by allowing the derivation of optimal "licensing" protocols for obtaining therapeutically efficient MSC.
Collapse
Affiliation(s)
- Bruno Sangiorgi
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | | | - Josiane Lilian Dos Santos Schiavinato
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | - Vitor Leão
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Haddad
- Faculdade de Ceilândia, Universidade de Brasília, Brasília, Brazil
| | | | - Vitor Marcel Faça
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil
| | | | - Dimas Tadeu Covas
- Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | - Marco Antônio Zago
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Alexandre Panepucci
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
31
|
Feng X, Lin Z, Sun W, Hollinger MK, Desierto MJ, Keyvanfar K, Malide D, Muranski P, Chen J, Young NS. Rapamycin is highly effective in murine models of immune-mediated bone marrow failure. Haematologica 2017; 102:1691-1703. [PMID: 28729300 PMCID: PMC5622853 DOI: 10.3324/haematol.2017.163675] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/12/2017] [Indexed: 12/14/2022] Open
Abstract
Acquired aplastic anemia, the prototypical bone marrow failure disease, is characterized by pancytopenia and marrow hypoplasia. Most aplastic anemia patients respond to immunosuppressive therapy, usually with anti-thymocyte globulin and cyclosporine, but some relapse on cyclosporine withdrawal or require long-term administration of cyclosporine to maintain blood counts. In this study, we tested efficacy of rapamycin as a new or alternative treatment in mouse models of immune-mediated bone marrow failure. Rapamycin ameliorated pancytopenia, improved bone marrow cellularity, and extended animal survival in a manner comparable to the standard dose of cyclosporine. Rapamycin effectively reduced Th1 inflammatory cytokines interferon-γ and tumor necrosis factor-α, increased the Th2 cytokine interleukin-10, stimulated expansion of functional regulatory T cells, eliminated effector CD8+ T cells (notably T cells specific to target cells bearing minor histocompatibility antigen H60), and preserved hematopoietic stem and progenitor cells. Rapamycin, but not cyclosporine, reduced the proportion of memory and effector T cells and maintained a pool of naïve T cells. Cyclosporine increased cytoplasmic nuclear factor of activated T-cells-1 following T-cell receptor stimulation, whereas rapamycin suppressed phosphorylation of two key signaling molecules in the mammalian target of rapamycin pathway, S6 kinase and protein kinase B. In summary, rapamycin was an effective therapy in mouse models of immune-mediated bone marrow failure, acting through different mechanisms to cyclosporine. Its specific expansion of regulatory T cells and elimination of clonogenic CD8+ effectors support its potential clinical utility in the treatment of aplastic anemia.
Collapse
Affiliation(s)
- Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zenghua Lin
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Hematology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Wanling Sun
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Maile K Hollinger
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marie J Desierto
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Keyvan Keyvanfar
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniela Malide
- Light Microscopy Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pawel Muranski
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jichun Chen
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
32
|
Weng L, Cao X, Han L, Zhao H, Qiu S, Yan Y, Wang X, Chen X, Zheng W, Xu X, Gao Y, Chen Y, Li J, Yang Y, Xu Y. Association of increased Treg and Th17 with pathogenesis of moyamoya disease. Sci Rep 2017; 7:3071. [PMID: 28596558 PMCID: PMC5465197 DOI: 10.1038/s41598-017-03278-8] [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: 10/31/2016] [Accepted: 04/26/2017] [Indexed: 01/03/2023] Open
Abstract
Immuno-inflammation has been shown to play a pivotal role in the pathogenesis of moyamoya disease (MMD). However, how did circulating Treg/Th17 cells involve in MMD patients remains unclear. 26 MMD, 21 atherothrombotic stroke, and 32 healthy controls were enrolled in this study. MMD patients have a significantly higher percentage of circulating Treg and Th17 cells as well as their dominantly secreting cytokines than other groups (P < 0.0001), whereas no difference was found in the ratio of Treg/Th17 between patients in MMD and atherothrombotic stroke group or control subjects (P = 0.244). However, the increased Treg in MMD patients which were enriched with FrIII Treg cells had deficient suppressive functions (P = 0.0017) compared to healthy volunteers. There was a positive correlation between Treg or TGF-β and MMD Suzuki’s stage. And the level of circulating Treg was as an independent factor associated with MMD stage. Besides, TGF-β was also correlated with the increased expression of VEGF in MMD patients. Our findings indicated an important involvement of circulating Treg in the pathogenic development of MMD and TGF-β in Treg induced VEGF.
Collapse
Affiliation(s)
- Leihua Weng
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Departments of Neurology, Affiliated Zhongshan Hospital of Xiamen University, 201 Hubinnan Road, Xiamen, 361004, China
| | - Xiang Cao
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Lijuan Han
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Haoran Zhao
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Shuwei Qiu
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Yaping Yan
- College of Cife Sciences, Shanxi Normal University, Xian, P. R. China
| | - Xiaoying Wang
- Departments of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xiangyan Chen
- Departments of Medicine and Therapeutics, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Weihong Zheng
- Departments of Neurology, Affiliated Zhongshan Hospital of Xiamen University, 201 Hubinnan Road, Xiamen, 361004, China
| | - Xin Xu
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Yuanyuan Gao
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Yan Chen
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Jie Li
- Department of Neurology, Affiliated Yixing People's Hospital of Jiangsu University, Yixing, Jiangsu, P. R. China
| | - Yongbo Yang
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China. .,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China.
| | - Yun Xu
- Department of Neurology and Neurosurgery, Drum Tower Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China. .,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China. .,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, P. R. China.
| |
Collapse
|
33
|
Zwang NA, Leventhal JR. Cell Therapy in Kidney Transplantation: Focus on Regulatory T Cells. J Am Soc Nephrol 2017; 28:1960-1972. [PMID: 28465379 DOI: 10.1681/asn.2016111206] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Renal transplantation is the renal replacement modality of choice for suitable candidates with advanced CKD or ESRD. Prevention of rejection, however, requires treatment with nonspecific pharmacologic immunosuppressants that carry both systemic and nephrologic toxicities. Use of a patient's own suppressive regulatory T cells (Tregs) is an attractive biologic approach to reduce this burden. Here, we review the immunologic underpinnings of Treg therapy and technical challenges to developing successful cell therapy. These issues include the selection of appropriate Treg subsets, ex vivo Treg expansion approaches, how many Tregs to administer and when, and how to care for patients after Treg administration.
Collapse
Affiliation(s)
| | - Joseph R Leventhal
- Comprehensive Transplant Center, Northwestern Memorial Hospital, Chicago, Illinois
| |
Collapse
|
34
|
Karasartova D, Gazi U, Tosun O, Gureser AS, Sahiner IT, Dolapci M, Ozkan AT. Anti-Pneumococcal Vaccine-Induced Cellular Immune Responses in Post-Traumatic Splenectomized Individuals. J Clin Immunol 2017; 37:388-396. [PMID: 28488145 DOI: 10.1007/s10875-017-0397-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/24/2017] [Indexed: 11/25/2022]
Abstract
PURPOSE Splenectomy is associated with increased risk of overwhelming post-splenectomy infections despite proper anti-pneumococcal vaccination. As most studies concentrated on vaccination-induced humoral immunity, the cellular immune responses triggered in splenectomized patients are not yet well studied. The present study aims to investigate this area as it can contribute to the development of more effective vaccination strategies. METHODS Five healthy and 14 splenectomized patients were vaccinated with pneumococcal conjugate polysaccharide vaccine (PCV) followed by pneumococcal polysaccharide vaccine according to the guidelines established by Advisory Committee on Immunization Practices. PBMC samples collected 0, 8, and 12 weeks after PCV immunization were in vitro stimulated with PCV. Levels of lymphoproliferation, TH cell differentiation, and cytokine release were assessed by carboxyfluorescein succinimidyl ester labeling, intracellular cytokine staining, and ELISA, respectively. RESULTS While TH1-dominated immune response was detected in both groups, asplenic individuals generated significantly lower levels of TH1 cells following in vitro stimulation. Similarly, levels of IFN-γ, IL-4, and IL-17 release and lymphoproliferation were significantly lower in asplenic patients. CONCLUSIONS According to our data, splenectomy negatively influences the levels of PCV-induced lymphoproliferation, TH1 differentiation, and cytokine release. Besides, PCV failed to induce TH17-dominant immune response which is crucial for protection against extracellular pathogens.
Collapse
Affiliation(s)
- Djursun Karasartova
- Department of Medical Microbiology, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Umut Gazi
- Department of Medical Microbiology and Clinic Microbiology, Faculty of Medicine, Near East University, Near East Boulevard, Nicosia, Cyprus.
| | - Ozgur Tosun
- Department of Biostatistics, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Ayse S Gureser
- Department of Medical Microbiology, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Ibrahim T Sahiner
- Department of General Surgery, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Mete Dolapci
- Department of General Surgery, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Aysegul T Ozkan
- Department of Medical Microbiology, Faculty of Medicine, Hitit University, Corum, Turkey
| |
Collapse
|
35
|
Park JH, Jang HR, Kim DH, Kwon GY, Lee JE, Huh W, Choi SJ, Oh W, Oh HY, Kim YG. Early, but not late, treatment with human umbilical cord blood-derived mesenchymal stem cells attenuates cisplatin nephrotoxicity through immunomodulation. Am J Physiol Renal Physiol 2017; 313:F984-F996. [PMID: 28356286 DOI: 10.1152/ajprenal.00097.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/13/2017] [Accepted: 03/23/2017] [Indexed: 11/22/2022] Open
Abstract
Preemptive treatment with mesenchymal stem cells (MSCs) can attenuate cisplatin-induced acute kidney injury (AKI). However, it is uncertain whether MSC treatment after the development of renal dysfunction prevents AKI progression or if MSC immunomodulatory properties contribute to MSC therapy. In this study, human umbilical cord blood (hUCB)-derived MSCs were used to compare the effects and mechanisms of early and late MSC therapy in a murine model. After cisplatin injection into C57BL/6 mice, hUCB-MSCs were administered on day 1 (early treatment) or day 3 (late treatment). With early treatment, cisplatin nephrotoxicity was attenuated as evidenced by decreased blood urea nitrogen (BUN) and reduced apoptosis and tubular injury scores on day 3 Early treatment resulted in downregulation of intrarenal monocyte chemotactic protein-1 and IL-6 expression and upregulation of IL-10 and VEGF expression. Flow cytometric analysis showed similar populations of infiltrated immune cells in both groups; however, regulatory T-cell (Treg) infiltration was 2.5-fold higher in the early treatment group. The role of Tregs was confirmed by the blunted effect of early treatment on renal injury after Treg depletion. In contrast, late treatment (at a time when BUN levels were 2-fold higher than baseline levels) showed no renoprotective effects on day 6 Neither the populations of intrarenal infiltrating immune cells (including Tregs) nor cytokine expression levels were affected by late treatment. Our results suggest that early MSC treatment attenuates renal injury by Treg induction and immunomodulation, whereas a late treatment (i.e., after the development of renal dysfunction) does not prevent AKI progression or alter the intrarenal inflammatory micromilieu.
Collapse
Affiliation(s)
- Ji Hyeon Park
- Division of Nephrology, Department of Medicine, Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Medicine, National Police Hospital, Seoul, Korea; and
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do Hee Kim
- Division of Nephrology, Department of Medicine, Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Ghee Young Kwon
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Eun Lee
- Division of Nephrology, Department of Medicine, Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Wooseong Huh
- Division of Nephrology, Department of Medicine, Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, R&D Center, Medipost Company, Gyeonggi-do, Korea
| | - Wonil Oh
- Biomedical Research Institute, R&D Center, Medipost Company, Gyeonggi-do, Korea
| | - Ha Young Oh
- Division of Nephrology, Department of Medicine, Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoon-Goo Kim
- Division of Nephrology, Department of Medicine, Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea;
| |
Collapse
|
36
|
Kerstein A, Schüler S, Cabral-Marques O, Fazio J, Häsler R, Müller A, Pitann S, Moosig F, Klapa S, Haas C, Kabelitz D, Riemekasten G, Wolters S, Lamprecht P. Environmental factor and inflammation-driven alteration of the total peripheral T-cell compartment in granulomatosis with polyangiitis. J Autoimmun 2017; 78:79-91. [DOI: 10.1016/j.jaut.2016.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/19/2016] [Accepted: 12/24/2016] [Indexed: 10/20/2022]
|
37
|
Baatjes AJ, Smith SG, Watson R, Howie K, Murphy D, Larché M, Denburg JA, Inman MD, O'Byrne PM. T regulatory cell phenotypes in peripheral blood and bronchoalveolar lavage from non-asthmatic and asthmatic subjects. Clin Exp Allergy 2016; 45:1654-62. [PMID: 26177872 DOI: 10.1111/cea.12594] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 05/08/2015] [Accepted: 06/03/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND An unresolved issue in T regulatory cells' cell biology is the lack of consensus on phenotypic markers that accurately define the natural Treg (nTreg) population. OBJECTIVES To examine nTreg frequency and functional capacity in healthy controls and their frequency in asthmatic subjects using three different phenotypic strategies. We hypothesized that phenotypically different nTreg are quantitatively and functionally different. METHODS Thirty-four healthy, non-asthmatic and 17 asthmatic subjects were studied. Three nTreg phenotypes were defined as follows: nTreg1 (CD4(+) CD25(+) Foxp3(+) ), nTreg2 (CD4(+) CD25(+) CD127(low) Foxp3(+) ), and nTreg3 (CD4(+) CD25(high) Foxp3(+) ). The flow cytometric determination of nTreg frequency in peripheral blood (PB) and bronchoalveolar lavage (BAL) was performed using fluorescently labelled antibodies. Peripheral blood nTreg functional capacity was assessed using a CFSE-based suppression assay. RESULTS There was a significantly lower frequency of PB nTreg3 compared to nTreg2 and nTreg1 (P < 0.05). Both nTreg2 and nTreg3 had a significantly greater suppressive capacity than nTreg1 at T responder (Tresp) to nTreg ratios of 16 : 1 up to 1 : 1 (P < 0.01). Asthmatics exhibited a significantly lower PB nTreg3 and nTreg1 frequency than healthy controls (P < 0.05). There were no differences between healthy controls and asthmatic subjects when comparing BAL nTreg frequency. CONCLUSIONS AND CLINICAL RELEVANCE Phenotypically different nTreg subsets are quantitatively and functionally different and are variably observed in asthma. The CD4(+) CD25(high) Foxp3(+) phenotype was the least frequent, but demonstrated the greatest suppression, and was significantly lower in PB of asthmatic subjects. Consequently, it is imperative that nTreg phenotypes be clearly defined and that the interpretation of their frequency and function be phenotype specific.
Collapse
Affiliation(s)
- A J Baatjes
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - S G Smith
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - R Watson
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - K Howie
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - D Murphy
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - M Larché
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - J A Denburg
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - M D Inman
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - P M O'Byrne
- Firestone Institute of Respiratory Health and the Department of Medicine, Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
38
|
Luc JGY, Paulin R, Zhao JY, Freed DH, Michelakis ED, Nagendran J. 2-Methoxyestradiol: A Hormonal Metabolite Modulates Stimulated T-Cells Function and proliferation. Transplant Proc 2016; 47:2057-66. [PMID: 26293097 DOI: 10.1016/j.transproceed.2015.05.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 05/09/2015] [Accepted: 05/15/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND 2-Methoxyestradiol (2ME2) is an endogenous metabolite of estrogen that is nonestrogenic and has been studied in cancer as an antimitotic agent that is beneficial by its selectivity for cancer cells without toxicity to nonmalignant cells. Because the effect of 2ME2 in a transplant rejection setting remains unknown, we hypothesized that 2ME2 can inhibit stimulated T-cell function. METHODS Human peripheral blood mononuclear cells (PBMCs) were cultured and pretreated with 2ME2 before stimulation. The cultured medium was collected for enzyme-linked immunosorbent assays, and whole-cell lysates were collected for Western immunoblotting. Proliferation and apoptosis assays were performed and analyzed by means of flow cytometry. RESULTS Tumor necrosis factor -α and interferon-γ cytokine production in 2ME2-treated stimulated PBMCs were modestly reduced relative to control samples. T-cell proliferation was blunted by treatment with 2ME2, and a decrease in apoptosis correlated with a decrease in caspase-9 activity. Additionally, 2ME2 was able to block stress-induced senescence caused by stimulation of T-cells. CONCLUSIONS 2ME2 is a hormone-based therapy that blunts stimulated T-cell proliferation and does not induce apoptosis or stress-induced senescence. Stimulated T-cells treated with 2ME2 are still able to produce normal levels of cytokines. Therefore, 2ME2 may lead to an oral immunomodulatory adjunct therapy with a low side effect profile for individuals undergoing transplantation.
Collapse
Affiliation(s)
- J G Y Luc
- Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - R Paulin
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - J Y Zhao
- Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - D H Freed
- Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada; Alberta Transplant Institute, Li Ka Shing Centre for Health Research, Edmonton, Alberta, Canada; Canadian National Transplant Research Program, Canada
| | - E D Michelakis
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - J Nagendran
- Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada; Alberta Transplant Institute, Li Ka Shing Centre for Health Research, Edmonton, Alberta, Canada; Canadian National Transplant Research Program, Canada.
| |
Collapse
|
39
|
van den Hout MFCM, Sluijter BJR, Santegoets SJAM, van Leeuwen PAM, van den Tol MP, van den Eertwegh AJM, Scheper RJ, de Gruijl TD. Local delivery of CpG-B and GM-CSF induces concerted activation of effector and regulatory T cells in the human melanoma sentinel lymph node. Cancer Immunol Immunother 2016; 65:405-15. [PMID: 26935057 PMCID: PMC4826413 DOI: 10.1007/s00262-016-1811-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 02/14/2016] [Indexed: 12/11/2022]
Abstract
Impaired immune effector functions in the melanoma sentinel lymph node (SLN) may allow for early metastatic events. In an effort to determine the optimal way to strengthen immune defenses, 28 clinical stage I-II melanoma patients were randomized in a 3-arm Phase II study to receive, prior to excision and sampling of the SLN, i.d. injections of saline or low-dose CpG-B (CpG), alone or combined with GM-CSF (GM), around the melanoma excision site. We previously described the combined administration of these DC-targeting agents to result in activation and recruitment of potentially cross-presenting BDCA3(+) DCs to the SLN. In this report we describe the effects on effector and regulatory T and NK cell subsets. Local low-dose CpG administration resulted in lower CD4/CD8 ratios, Th1 skewing, increased frequencies of melanoma-specific CD8(+) T cells and possible recruitment of effector NK cells, irrespective of GM co-administration. These immune-potentiating effects were counterbalanced by increased IL-10 production by T cells and significantly higher levels of FoxP3 and CTLA4 in regulatory T cells (Tregs) with correspondingly higher suppressive activity in the SLN. Notably, CpG ± GM-administered patients showed significantly lower numbers of SLN metastases (saline: 4/9, CpG + GM: 1/9, CpG: 0/10, p = 0.04). These findings indicate that i.d. delivery of low-dose CpG ± GM potentially arms the SLN of early-stage melanoma patients against metastatic spread, but that antitumor efficacy may be further boosted by counteracting the collateral activation of Tregs.
Collapse
Affiliation(s)
- Mari F C M van den Hout
- Department of Pathology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Berbel J R Sluijter
- Department of Surgical Oncology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Saskia J A M Santegoets
- Department of Medical Oncology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, De Boelelaan 1117, Room CCA 2.44, 1081 HV, Amsterdam, The Netherlands
| | - Paul A M van Leeuwen
- Department of Surgical Oncology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - M Petrousjka van den Tol
- Department of Surgical Oncology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, De Boelelaan 1117, Room CCA 2.44, 1081 HV, Amsterdam, The Netherlands
| | - Rik J Scheper
- Department of Pathology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, De Boelelaan 1117, Room CCA 2.44, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Akimova T, Levine MH, Beier UH, Hancock WW. Standardization, Evaluation, and Area-Under-Curve Analysis of Human and Murine Treg Suppressive Function. Methods Mol Biol 2016; 1371:43-78. [PMID: 26530794 DOI: 10.1007/978-1-4939-3139-2_4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
FOXP3+ T-regulatory (Treg) cells have important roles in immune homeostasis, and alterations in their number and function can predispose to diseases ranging from autoimmunity to allograft rejection and tumor growth. Reliable identification of human Tregs remains a persistent problem due to a lack of specific markers. The most definitive Treg characterization currently involves combined assessment of phenotypic, epigenetic and functional parameters, with the latter typically involving in vitro Treg suppression assays. Unfortunately, suppression assays are frequently performed using differing methods and readouts, limiting comparisons between studies. We provide a perspective on our experience with human and murine Treg suppression assay conditions, including Treg data obtained in clinical transplant studies, Tregs isolated from healthy donors and treated with epigenetically active compounds, and Tregs from standard murine strains (C57BL/6 and BALB/c). We provide detailed descriptions and illustrations of typical problems, shortcomings and troubleshooting; describe new modifications and approaches; and present a new method for calculation of suppressive assay data using a modified area-under-curve (AUC) method. This method allows us to directly compare Treg suppressive function between multiple patients (such as in clinical transplant studies), to reliably track changes in Treg function from the same person over time, or compare effects of Treg-modulating compounds tested with different healthy donors Tregs in separate or combined experimental settings.
Collapse
Affiliation(s)
- Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine and Biesecker Center for Pediatric Liver Disease, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Matthew H Levine
- Department of Surgery, Penn Transplant Institute, Hospital of the University of Pennsylvania and University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ulf H Beier
- Division of Nephrology, Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wayne W Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine and Biesecker Center for Pediatric Liver Disease, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
41
|
Li J, Du X, Shi H, Deng K, Chi H, Tao W. Mammalian Sterile 20-like Kinase 1 (Mst1) Enhances the Stability of Forkhead Box P3 (Foxp3) and the Function of Regulatory T Cells by Modulating Foxp3 Acetylation. J Biol Chem 2015; 290:30762-70. [PMID: 26538561 PMCID: PMC4692206 DOI: 10.1074/jbc.m115.668442] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 11/01/2015] [Indexed: 11/06/2022] Open
Abstract
Regulatory T cells (Tregs) play crucial roles in maintaining immune tolerance. The transcription factor Foxp3 is a critical regulator of Treg development and function, and its expression is regulated at both transcriptional and post-translational levels. Acetylation by lysine acetyl transferases/lysine deacetylases is one of the main post-translational modifications of Foxp3, which regulate Foxp3's stability and transcriptional activity. However, the mechanism(s) by which the activities of these lysine acetyl transferases/lysine deacetylases are regulated to preserve proper Foxp3 acetylation during Treg development and maintenance of Treg function remains to be determined. Here we report that Mst1 can enhance Foxp3 stability, its transcriptional activity, and Treg function by modulating the Foxp3 protein at the post-translational level. We discovered that Mst1 could increase the acetylation of Foxp3 by inhibiting Sirt1 activity, which requires the Mst1 kinase activity. We also found that Mst1 could attenuate Sirt1-mediated deacetylation of Foxp3 through directly interacting with Foxp3 to prevent or interfere the interaction between Sirt1 and Foxp3. Therefore, Mst1 can regulate Foxp3 stability in kinase-dependent and kinase-independent manners. Finally, we showed that treatment of Mst1(-/-) Tregs with Ex-527, a Sirt1-specific inhibitor, partially restored the suppressive function of Mst1(-/-) Tregs. Our studies reveal a novel mechanism by which Mst1 enhances Foxp3 expression and Treg function at the post-translational level.
Collapse
Affiliation(s)
- Jiang Li
- the Obstetrics and Gynecology Hospital and Institute of Reproduction and Development, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai 200011, China, and From the Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xingrong Du
- From the Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China, the Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Hao Shi
- From the Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Kejing Deng
- From the Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Hongbo Chi
- the Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Wufan Tao
- the Obstetrics and Gynecology Hospital and Institute of Reproduction and Development, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai 200011, China, and From the Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China,
| |
Collapse
|
42
|
Moon BI, Kim TH, Seoh JY. Functional Modulation of Regulatory T Cells by IL-2. PLoS One 2015; 10:e0141864. [PMID: 26529512 PMCID: PMC4631326 DOI: 10.1371/journal.pone.0141864] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/14/2015] [Indexed: 01/31/2023] Open
Abstract
The suppressive function of regulatory T cells (Tregs) is critical to the maintenance of immune homeostasis in vivo and yet, the specific identification of Tregs by phenotypic markers is not perfect. Tregs were originally identified in the CD4+CD25+ fraction of T cells, but FoxP3 expression was later included as an additional marker of Tregs as FoxP3 expression was identified as being critical to the development and function of these cells. Intracellular expression of FoxP3 makes it difficult in using to isolate live and not permeabilized cells for functional assays. As such CD4+CD25+ fraction is still frequently used for functional assays of Tregs. Although, the CD4+CD25+ fraction substantially overlaps with the FoxP3+ fraction, the minor mismatch between CD4+CD25+ and FoxP3+ fractions may confound the functional characteristics of Tregs. In this study, we isolated CD4+FoxP3+ as well as CD4+CD25+ fractions from Foxp3 knock-in mice, and compared their proliferative and suppressive activity in the presence or absence of various concentrations of IL-2. Our results showed comparable patterns of proliferative and suppressive responses for both fractions, except that contrary to the CD4+CD25+ fraction the FoxP3+ fraction did not proliferate in an autocrine fashion even in response to a strong stimulation. In presence of exogenous IL-2, both CD4+CD25+ and CD4+FoxP3+ fractions were more sensitive than the CD4+CD25- responder cells in proliferative responsiveness. In addition, a low dose IL-2 enhanced whereas a high dose abrogated the suppressive activities of the CD4+CD25+ and CD4+FoxP3+ fractions. These results may provide an additional understanding of the characteristics of the various fractions of isolated Tregs based on phenotype and function and the role of varying levels of exogenous IL-2 on the suppressive activity of these cells.
Collapse
Affiliation(s)
- Byung-In Moon
- Department of Surgery, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Tae Hun Kim
- Department of Internal medicine, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Ju-Young Seoh
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
43
|
Eckl‐Dorna J, Campana R, Valenta R, Niederberger V. Poor association of allergen-specific antibody, T- and B-cell responses revealed with recombinant allergens and a CFSE dilution-based assay. Allergy 2015; 70:1222-9. [PMID: 26043182 PMCID: PMC4949646 DOI: 10.1111/all.12661] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND The adaptive immunity underlying allergy comprises two components, the allergen-specific antibody (i.e. IgE, IgG) and the T-cell response. These two components are responsible for different disease manifestations and can be targeted by different therapeutic approaches. Here, we investigated the association of allergen-specific antibody and T- as well as B-cell responses in pollen-allergic patients using recombinant (r) major birch pollen allergen rBet v 1 and major timothy grass pollen allergen rPhl p 5 as defined antigens. METHODS Allergen-specific IgE and IgG antibody responses were determined by ELISA, and allergen-specific T- and B-cell responses were measured in peripheral blood mononuclear cells using a carboxyfluorescein-diacetate-succinimidylester (CFSE) dilution assay. RESULTS CFSE staining in combination with T-cell- and B-cell-specific gating allowed discriminating between allergen-specific T-cell and B-cell responses. Interestingly, we identified patients where mainly T cells and others where mainly B cells proliferated in response to allergen stimulation. No association between the level of allergen-specific Ig responses and B- or T-cell proliferation was observed. CONCLUSION Purified recombinant allergens in conjunction with CFSE staining allow the dissection of allergen-specific B- and T-cell responses. The dissociation of allergen-specific antibody, and B- and T-cell responses may explain the occurrence of selective IgE- and T-cell-mediated manifestations of allergic inflammation and may be important for the development of diagnostic and therapeutic strategies selectively targeting B cells and T cells.
Collapse
Affiliation(s)
- J. Eckl‐Dorna
- Department of Otorhinolaryngology Medical University of Vienna Vienna Austria
| | - R. Campana
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - R. Valenta
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - V. Niederberger
- Department of Otorhinolaryngology Medical University of Vienna Vienna Austria
| |
Collapse
|
44
|
Dhaeze T, Peelen E, Hombrouck A, Peeters L, Van Wijmeersch B, Lemkens N, Lemkens P, Somers V, Lucas S, Broux B, Stinissen P, Hellings N. Circulating Follicular Regulatory T Cells Are Defective in Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2015; 195:832-40. [DOI: 10.4049/jimmunol.1500759] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/18/2015] [Indexed: 12/29/2022]
|
45
|
Abstract
Chikungunya virus (CHIKV) infection is a reemerging pandemic human arboviral disease. CD4+ T cells were previously shown to contribute to joint inflammation in the course of CHIKV infection in mice. The JES6-1 anti-IL-2 antibody selectively expands mouse regulatory T cells (Tregs) by forming a complex with IL-2. In this study, we show that the IL-2 JES6-1-mediated expansion of Tregs ameliorates CHIKV-induced joint pathology. It does so by inhibiting the infiltration of CD4+ T cells due to the induction of anergy in CHIKV-specific CD4+ effector T cells. These findings suggest that activation of Tregs could also become an alternative approach to control CHIKV-mediated disease.
IMPORTANCE Chikungunya virus (CHIKV) has reemerged as a pathogen of global significance. Patients infected with CHIKV suffer from incapacitating joint pain that severely affects their daily functioning. Despite the best efforts, treatment is still inadequate. While T cell-mediated immunopathology in CHIKV infections has been reported, the role of regulatory T cells (Tregs) has not been explored. The JES6-1 anti-interleukin 2 (IL-2) antibody has been demonstrated to selectively expand mouse Tregs by forming a complex with IL-2. We reveal here that IL-2 JES6-1-mediated expansion of Tregs ameliorates CHIKV-induced joint pathology in mice by neutralizing virus-specific CD4+ effector T (Teff) cells. We show that this treatment abrogates the infiltration of pathogenic CD4+ T cells through induction of anergy in CHIKV-specific CD4+ Teff cells. This is the first evidence where the role of Tregs is demonstrated in CHIKV pathogenesis, and its expansion could control virus-mediated immunopathology.
Collapse
|
46
|
ElEssawy B, Li XC. Type 1 diabetes and T regulatory cells. Pharmacol Res 2015; 98:22-30. [PMID: 25959211 DOI: 10.1016/j.phrs.2015.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 12/11/2022]
Abstract
T-regulatory cells (Tregs) play a fundamental role in the creation and maintenance of peripheral tolerance. Deficits in the numbers and/or function of Tregs may be an underlying cause of human autoimmune diseases including type 1 Diabetes Mellitus (T1D), whereas an over-abundance of Tregs can hinder immunity against cancer or pathogens. The importance of Tregs in the control of autoimmunity is well established in a variety of experimental animal models. In mice, manipulating the numbers and/or function of Tregs can decrease pathology in a wide range of contexts, including autoimmunity and it is widely assumed that similar approaches will be possible in humans. T1D, the most prevalent human autoimmune disease, has been a focus of interventions either through direct and indirect in vivo proliferations or through adoptive transfer of the in vitro generated antigen specific and non specific Treg. Some challenges still need to be addressed, including a more specific phenotype marker for Tregs; the reproducibility of satisfactory animal results in human and the reconcile of discrepancies between in vitro and in vivo studies. In this article, we will highlight the role of Tregs in autoimmune disease in general with a special focus on T1D, highlighting progress made and challenges ahead in developing Treg-based therapies.
Collapse
Affiliation(s)
| | - Xian C Li
- Immunobiology & Transplantation Research, Houston Methodist Hospital, Texas Medical Center, 6670 Bertner Avenue, R7-211, Houston, TX 77030, United States.
| |
Collapse
|
47
|
Nakano S, Morimoto S, Suzuki S, Tsushima H, Yamanaka K, Sekigawa I, Takasaki Y. Immunoregulatory role of IL-35 in T cells of patients with rheumatoid arthritis. Rheumatology (Oxford) 2015; 54:1498-506. [PMID: 25731770 DOI: 10.1093/rheumatology/keu528] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE IL-35 is the most recently identified member of the IL-12 family. It consists of EBV-induced gene 3 (EBI3) and IL-12α chain p35. We investigated whether IL-35 enhances the in vitro immunosuppressive function of peripheral blood isolated from patients with RA. METHODS Peripheral blood was harvested from 17 active and 10 inactive RA patients and IL-35 concentrations were quantified using an ELISA. An expression vector containing IL-35 with a FLAG tag at the carboxyl-terminus was constructed by covalently linking EBI3 and IL-12α (p35). The function of IL-35 was then evaluated in a suppression assay using T cells isolated from human RA patients with CD2, CD3 and CD28 antibodies. RESULTS Serum IL-35 levels and the number of Treg were decreased significantly in patients with active RA. There was a significant correlation between serum IL-35 and the 28-joint DAS with ESR (DAS28-ESR) in patients with active RA. IL-35 treatment enhanced the regulatory function, suppressing the levels of inflammatory cytokines such as IL-17 and IFN-γ and the cellular growth of effector T cells stimulated by conjugation with CD2, CD3 and CD28. CONCLUSION These data revealed that IL-35 might suppress T cell activation during the peripheral immune responses of RA. Therefore our data suggest that IL-35 might have multiple therapeutic targets.
Collapse
Affiliation(s)
- Souichiro Nakano
- Department of Rheumatology and Internal Medicine, Juntendo University School of Medicine, Tokyo,
| | - Shinji Morimoto
- Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba and
| | - Satoshi Suzuki
- Department of Rheumatology and Internal Medicine, Juntendo University School of Medicine, Tokyo
| | - Hiroshi Tsushima
- Department of Rheumatology and Internal Medicine, Juntendo University School of Medicine, Tokyo
| | - Kenjiro Yamanaka
- Department of Rheumatology and Internal Medicine, Sasaki Foundation, Tokyo, Japan
| | - Iwao Sekigawa
- Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba and
| | - Yoshinari Takasaki
- Department of Rheumatology and Internal Medicine, Juntendo University School of Medicine, Tokyo
| |
Collapse
|
48
|
Morande PE, Borge M, Abreu C, Galletti J, Zanetti SR, Nannini P, Bezares RF, Pantano S, Dighiero G, Oppezzo P, Gamberale R, Giordano M. Surface localization of high-mobility group nucleosome-binding protein 2 on leukemic B cells from patients with chronic lymphocytic leukemia is related to secondary autoimmune hemolytic anemia. Leuk Lymphoma 2015; 56:1115-22. [DOI: 10.3109/10428194.2014.957205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
49
|
Valdez-Ortiz R, Bestard O, Llaudó I, Franquesa M, Cerezo G, Torras J, Herrero-Fresneda I, Correa-Rotter R, Grinyó JM. Induction of suppressive allogeneic regulatory T cells via rabbit antithymocyte polyclonal globulin during homeostatic proliferation in rat kidney transplantation. Transpl Int 2014; 28:108-19. [PMID: 25208307 DOI: 10.1111/tri.12448] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/19/2014] [Accepted: 09/04/2014] [Indexed: 11/27/2022]
Abstract
Experimental studies have shown that rabbit antithymocyte polyclonal globulin (ATG) can expand human CD4+CD25++Foxp3+ cells (Tregs). We investigated the major biological effects of a self-manufactured rabbit polyclonal anti-rat thymoglobulin (rATG) in vitro, as well as its effects on different peripheral T-cell subsets. Moreover, we evaluated the allogeneic suppressive capacity of rATG-induced Tregs in an experimental rat renal transplant model. Our results show that rATG has the capacity to induce apoptosis in T lymphocyte lymphocytes as a primary mechanism of T-cell depletion. Our in vivo studies demonstrated a rapid but transient cellular depletion of the main T cell subsets, directly proportional to the rATG dose used, but not of the effector memory T cells, which required significantly higher rATG doses. After rATG administration, we observed a significant proliferation of Tregs in the peripheral blood of transplanted rats, leading to an increase in the Treg/T effector ratio. Importantly, rATG-induced Tregs displayed a strong donor-specific suppressive capacity when assessed in an antigen-specific allogeneic co-culture. All of these results were associated with better renal graft function in rats that received rATG. Our study shows that rATG has the biological capacity immunomodulatory to promote a regulatory alloimmune milieu during post-transplant homeostatic proliferation.
Collapse
Affiliation(s)
- Rafael Valdez-Ortiz
- Laboratory of Experimental Nephrology, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Nephrology Department, Hospital General de México, Mexico City, México; Renal Transplant Unit, Department of Nephrology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Grant CR, Liberal R, Mieli-Vergani G, Vergani D, Longhi MS. Regulatory T-cells in autoimmune diseases: challenges, controversies and--yet--unanswered questions. Autoimmun Rev 2014; 14:105-16. [PMID: 25449680 DOI: 10.1016/j.autrev.2014.10.012] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 09/26/2014] [Indexed: 02/07/2023]
Abstract
Regulatory T cells (Tregs) are central to the maintenance of self-tolerance and tissue homeostasis. Markers commonly used to define human Tregs in the research setting include high expression of CD25, FOXP3 positivity and low expression/negativity for CD127. Many other markers have been proposed, but none unequivocally identifies bona fide Tregs. Tregs are equipped with an array of mechanisms of suppression, including the modulation of antigen presenting cell maturation and function, the killing of target cells, the disruption of metabolic pathways and the production of anti-inflammatory cytokines. Treg impairment has been reported in a number of human autoimmune conditions and includes Treg numerical and functional defects and conversion into effector cells in response to inflammation. In addition to intrinsic Treg impairment, resistance of effector T cells to Treg control has been described. Discrepancies in the literature are common, reflecting differences in the choice of study participants and the technical challenges associated with investigating this cell population. Studies differ in terms of the methodology used to define and isolate putative regulatory cells and to assess their suppressive function. In this review we outline studies describing Treg frequency and suppressive function in systemic and organ specific autoimmune diseases, with a specific focus on the challenges faced when investigating Tregs in these conditions.
Collapse
Affiliation(s)
- Charlotte R Grant
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, London, United Kingdom; Faculty of Life Sciences and Medicine, King's College Hospital, London, United Kingdom.
| | - Rodrigo Liberal
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, London, United Kingdom; Faculty of Life Sciences and Medicine, King's College Hospital, London, United Kingdom.
| | - Giorgina Mieli-Vergani
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, London, United Kingdom; Faculty of Life Sciences and Medicine, King's College Hospital, London, United Kingdom.
| | - Diego Vergani
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, London, United Kingdom; Faculty of Life Sciences and Medicine, King's College Hospital, London, United Kingdom.
| | - Maria Serena Longhi
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, London, United Kingdom; Faculty of Life Sciences and Medicine, King's College Hospital, London, United Kingdom.
| |
Collapse
|