1
|
Choi S, Yoo SA, Ji KY, Jung DH, Lee S, Lee KG, Kim KM, Lee JY, Jung MA, Pyun BJ, Hur J, Choi JY, Rhee CK, Kim WU, Kim T. Asthma Alleviation by Ginsenoside Rb1 via Promotion of Treg Proliferation and Inflammatory T Cell Inhibition. Allergy 2025. [PMID: 40251907 DOI: 10.1111/all.16551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/09/2025] [Accepted: 02/20/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Regulatory T cells (Tregs) are living drugs with feasibility, tolerability, and therapeutic benefits. Although Tregs are linked to asthma prognosis through inflammation regulation, no therapeutic agents specifically designed to manage asthma by upregulating Tregs have been developed to date. METHODS We screened a library of 250 natural products using a cytometric bead array. Among the selected candidates, gRb1 was identified for further investigation. The effects of gRb1 on Treg and Th17 populations were evaluated in mouse asthma models and human PBMCs from both healthy donors and asthma patients using flow cytometry and cytokine analysis. RESULTS In inflammatory conditions, ginsenoside Rb1 (gRb1, a major ginseng component) increased IL-10- and TGF-β-expressing Treg populations and decreased the Th17 population; activated phospho-STAT5 and NFAT1 in Tregs; inhibited NFAT1 activation in conventional T cells (Tconvs); increased Treg proliferation and Tconv-Treg differentiation, inhibiting Tconv proliferation; and reduced inflammatory cytokine secretion by Tconvs. In asthma model mice, suppression of asthma symptoms by gRb1 was associated with elevated Treg and lower Th17, Th1, and Th2 counts. gRb1 treatment of stimulated PBMCs from patients with asthma and healthy donors increased IL-10- and TGF-β-expressing Treg populations and decreased IL-17A-, IL-22-, IFN-γ-, and TNF-α-expressing T-cell populations. CONCLUSIONS gRb1 alleviate asthma by shifting the Treg-inflammatory T cell balance. These findings suggest a strategy for enhancing Treg activity through treatment with gRb1. This may provide a novel therapeutic approach for asthma and related disorders.
Collapse
Affiliation(s)
- Susanna Choi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Seung-Ah Yoo
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kon-Young Ji
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Dong Ho Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Saseong Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kang-Gu Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ki-Myo Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul National University Boramae Hospital, Seoul, Republic of Korea
| | - Joo Young Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Myung-A Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Bo-Jeong Pyun
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Jung Hur
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| |
Collapse
|
2
|
Geels SN, Moshensky A, Sousa RS, Murat C, Bustos MA, Walker BL, Singh R, Harbour SN, Gutierrez G, Hwang M, Mempel TR, Weaver CT, Nie Q, Hoon DSB, Ganesan AK, Othy S, Marangoni F. Interruption of the intratumor CD8 + T cell:Treg crosstalk improves the efficacy of PD-1 immunotherapy. Cancer Cell 2024; 42:1051-1066.e7. [PMID: 38861924 PMCID: PMC11285091 DOI: 10.1016/j.ccell.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 02/28/2024] [Accepted: 05/14/2024] [Indexed: 06/13/2024]
Abstract
PD-1 blockade unleashes potent antitumor activity in CD8+ T cells but can also promote immunosuppressive T regulatory (Treg) cells, which may worsen the response to immunotherapy. Tumor-Treg inhibition is a promising strategy to improve the efficacy of checkpoint blockade immunotherapy; however, our understanding of the mechanisms supporting tumor-Tregs during PD-1 immunotherapy is incomplete. Here, we show that PD-1 blockade increases tumor-Tregs in mouse models of melanoma and metastatic melanoma patients. Mechanistically, Treg accumulation is not caused by Treg-intrinsic inhibition of PD-1 signaling but depends on an indirect effect of activated CD8+ T cells. CD8+ T cells produce IL-2 and colocalize with Tregs in mouse and human melanomas. IL-2 upregulates the anti-apoptotic protein ICOS on tumor-Tregs, promoting their accumulation. Inhibition of ICOS signaling before PD-1 immunotherapy improves control over immunogenic melanoma. Thus, interrupting the intratumor CD8+ T cell:Treg crosstalk represents a strategy to enhance the therapeutic efficacy of PD-1 immunotherapy.
Collapse
Affiliation(s)
- Shannon N Geels
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Alexander Moshensky
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Rachel S Sousa
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Claire Murat
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Matias A Bustos
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Santa Monica, CA, USA
| | - Benjamin L Walker
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Rima Singh
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Stacey N Harbour
- Department of Pathology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Giselle Gutierrez
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| | - Michael Hwang
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Thorsten R Mempel
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Casey T Weaver
- Department of Pathology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Qing Nie
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Dave S B Hoon
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Santa Monica, CA, USA
| | - Anand K Ganesan
- Department of Dermatology, University of California, Irvine, Irvine, CA, USA
| | - Shivashankar Othy
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Francesco Marangoni
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
3
|
Bader CS, Pavlova A, Lowsky R, Muffly LS, Shiraz P, Arai S, Johnston LJ, Rezvani AR, Weng WK, Miklos DB, Frank MJ, Tamaresis JS, Agrawal V, Bharadwaj S, Sidana S, Shizuru JA, Fernhoff NB, Putnam A, Killian S, Xie BJ, Negrin RS, Meyer EH. Single-center randomized trial of T-reg graft alone vs T-reg graft plus tacrolimus for the prevention of acute GVHD. Blood Adv 2024; 8:1105-1115. [PMID: 38091578 PMCID: PMC10907400 DOI: 10.1182/bloodadvances.2023011625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/27/2023] [Indexed: 02/29/2024] Open
Abstract
ABSTRACT Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for hematological malignancies for which graft-versus-host disease (GVHD) remains a major complication. The use of donor T-regulatory cells (Tregs) to prevent GVHD appears promising, including in our previous evaluation of an engineered graft product (T-reg graft) consisting of the timed, sequential infusion of CD34+ hematopoietic stem cells and high-purity Tregs followed by conventional T cells. However, whether immunosuppressive prophylaxis can be removed from this protocol remains unclear. We report the results of the first stage of an open-label single-center phase 2 study (NCT01660607) investigating T-reg graft in myeloablative HCT of HLA-matched and 9/10-matched recipients. Twenty-four patients were randomized to receive T-reg graft alone (n = 12) or T-reg graft plus single-agent GVHD prophylaxis (n = 12) to determine whether T-reg graft alone was noninferior in preventing acute GVHD. All patients developed full-donor myeloid chimerism. Patients with T-reg graft alone vs with prophylaxis had incidences of grade 3 to 4 acute GVHD of 58% vs 8% (P = .005) and grade 3 to 4 of 17% vs 0% (P = .149), respectively. The incidence of moderate-to-severe chronic GVHD was 28% in the T-reg graft alone arm vs 0% with prophylaxis (P = .056). Among patients with T-reg graft and prophylaxis, CD4+ T-cell-to-Treg ratios were reduced after transplantation, gene expression profiles showed reduced CD4+ proliferation, and the achievement of full-donor T-cell chimerism was delayed. This study indicates that T-reg graft with single-agent tacrolimus is preferred over T-reg graft alone for the prevention of acute GVHD. This trial was registered at www.clinicaltrials.gov as #NCT01660607.
Collapse
Affiliation(s)
- Cameron S. Bader
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Anna Pavlova
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Robert Lowsky
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| | - Lori S. Muffly
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Parveen Shiraz
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Sally Arai
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| | - Laura J. Johnston
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Andrew R. Rezvani
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Wen-Kai Weng
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| | - David B. Miklos
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Matthew J. Frank
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | | | - Vaibhav Agrawal
- Department of Hematology and Hematopoietic Stem Cell Transplantation, City of Hope, Duarte, CA
| | - Sushma Bharadwaj
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Surbhi Sidana
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Judith A. Shizuru
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | | | | | | | | | - Robert S. Negrin
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| | - Everett H. Meyer
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| |
Collapse
|
4
|
Geels SN, Moshensky A, Sousa RS, Walker BL, Singh R, Gutierrez G, Hwang M, Mempel TR, Nie Q, Othy S, Marangoni F. Interruption of the Intratumor CD8:Treg Crosstalk Improves the Efficacy of PD-1 Immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540889. [PMID: 37292782 PMCID: PMC10245792 DOI: 10.1101/2023.05.15.540889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
PD-1 blockade unleashes the potent antitumor activity of CD8 cells but can also promote immunosuppressive T regulatory (Treg) cells, which may worsen response to immunotherapy. Tumor Treg inhibition is a promising strategy to overcome therapeutic resistance; however, the mechanisms supporting tumor Tregs during PD-1 immunotherapy are largely unexplored. Here, we report that PD-1 blockade increases tumor Tregs in mouse models of immunogenic tumors, including melanoma, and metastatic melanoma patients. Unexpectedly, Treg accumulation was not caused by Treg-intrinsic inhibition of PD-1 signaling but instead depended on an indirect effect of activated CD8 cells. CD8 cells colocalized with Tregs within tumors and produced IL-2, especially after PD-1 immunotherapy. IL-2 upregulated the anti-apoptotic protein ICOS on tumor Tregs, causing their accumulation. ICOS signaling inhibition before PD-1 immunotherapy resulted in increased control of immunogenic melanoma. Thus, interrupting the intratumor CD8:Treg crosstalk is a novel strategy that may enhance the efficacy of immunotherapy in patients.
Collapse
|
5
|
Hirai T, Lin PY, Ramos TL, Simonetta F, Su LL, Picton LK, Baker J, Lohmeyer JK, Garcia KC, Negrin RS. IL-2 receptor engineering enhances regulatory T cell function suppressed by calcineurin inhibitor. Am J Transplant 2022; 22:3061-3068. [PMID: 36031344 PMCID: PMC10184573 DOI: 10.1111/ajt.17181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/27/2022] [Accepted: 08/15/2022] [Indexed: 01/25/2023]
Abstract
Clinical trials utilizing regulatory T cell (Treg) therapy in organ transplantation have shown promising results, however, the choice of a standard immunosuppressive regimen is still controversial. Calcineurin inhibitors (CNIs) are one of the most common immunosuppressants for organ transplantation, although they may negatively affect Tregs by inhibiting IL-2 production by conventional T cells. As a strategy to replace IL-2 signaling selectively in Tregs, we have introduced an engineered orthogonal IL-2 (ortho IL-2) cytokine/cytokine receptor (R) pair that specifically binds with each other but does not bind with their wild-type counterparts. Murine Tregs were isolated from recipients and retrovirally transduced with ortho IL-2Rβ during ex vivo expansion. Transduced Tregs (ortho Tregs) were transferred into recipient mice in a mixed hematopoietic chimerism model with tacrolimus administration. Ortho IL-2 treatment significantly increased the ortho IL-2Rβ(+) Treg population in the presence of tacrolimus without stimulating other T cell subsets. All the mice treated with tacrolimus plus ortho IL-2 achieved heart allograft tolerance, even after tacrolimus cessation, whereas those receiving tacrolimus treatment alone did not. These data demonstrate that Treg therapy can be adopted into a CNI-based regimen by utilizing cytokine receptor engineering.
Collapse
Affiliation(s)
- Toshihito Hirai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Po-Yu Lin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California, USA
| | - Teresa L. Ramos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California, USA
| | - Federico Simonetta
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California, USA
| | - Leon L. Su
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Lora K. Picton
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California, USA
| | - Juliane K. Lohmeyer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California, USA
| | - K. Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Robert S. Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
6
|
TCR-induced FOXP3 expression by CD8 + T cells impairs their anti-tumor activity. Cancer Lett 2022; 528:45-58. [PMID: 34973390 DOI: 10.1016/j.canlet.2021.12.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/09/2021] [Accepted: 12/25/2021] [Indexed: 11/23/2022]
Abstract
Adoptive cell transfer therapy using CD8+ T lymphocytes showed promising results eradicating metastatic malignancies. However, several regulatory mechanisms limit its efficacy. We studied the role of the expression of the transcription factor FOXP3 on CD8+ T cell function and anti-tumor immunity. Here we show that suboptimal T cell receptor stimulation of CD8+ T cells upregulates FOXP3 in vitro. Similarly, CD8 T cells transferred into tumor-bearing mice upregulate FOXP3 in vivo. Cell-intrinsic loss of FOXP3 by CD8+ T cells resulted in improved functionality after TCR stimulation and better antitumor responses in vivo. Inhibition of the FOXP3/NFAT interaction likewise improved CD8+ T cell functionality. Transcriptomic analysis of cells after TCR stimulation revealed an enrichment of genes implicated in the response to IFN-γ, IFN-α, inflammatory response, IL-6/JAK/STAT, G2M checkpoint and IL-2/STAT signaling in FOXP3-deficient CD8+ T cells with respect to FOXP3-wt CD8+ T cells. Our results suggest that transient expression of FOXP3 by CD8+ T cells in the tumor microenvironment restrains their anti-tumor activity, with clear implications for improving T cell responses during immunotherapy.
Collapse
|
7
|
K 2P18.1 translates T cell receptor signals into thymic regulatory T cell development. Cell Res 2022; 32:72-88. [PMID: 34702947 PMCID: PMC8547300 DOI: 10.1038/s41422-021-00580-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
It remains largely unclear how thymocytes translate relative differences in T cell receptor (TCR) signal strength into distinct developmental programs that drive the cell fate decisions towards conventional (Tconv) or regulatory T cells (Treg). Following TCR activation, intracellular calcium (Ca2+) is the most important second messenger, for which the potassium channel K2P18.1 is a relevant regulator. Here, we identify K2P18.1 as a central translator of the TCR signal into the thymus-derived Treg (tTreg) selection process. TCR signal was coupled to NF-κB-mediated K2P18.1 upregulation in tTreg progenitors. K2P18.1 provided the driving force for sustained Ca2+ influx that facilitated NF-κB- and NFAT-dependent expression of FoxP3, the master transcription factor for Treg development and function. Loss of K2P18.1 ion-current function induced a mild lymphoproliferative phenotype in mice, with reduced Treg numbers that led to aggravated experimental autoimmune encephalomyelitis, while a gain-of-function mutation in K2P18.1 resulted in increased Treg numbers in mice. Our findings in human thymus, recent thymic emigrants and multiple sclerosis patients with a dominant-negative missense K2P18.1 variant that is associated with poor clinical outcomes indicate that K2P18.1 also plays a role in human Treg development. Pharmacological modulation of K2P18.1 specifically modulated Treg numbers in vitro and in vivo. Finally, we identified nitroxoline as a K2P18.1 activator that led to rapid and reversible Treg increase in patients with urinary tract infections. Conclusively, our findings reveal how K2P18.1 translates TCR signals into thymic T cell fate decisions and Treg development, and provide a basis for the therapeutic utilization of Treg in several human disorders.
Collapse
|
8
|
Hernández Vásquez MN, Ulvmar MH, González-Loyola A, Kritikos I, Sun Y, He L, Halin C, Petrova TV, Mäkinen T. Transcription factor FOXP2 is a flow-induced regulator of collecting lymphatic vessels. EMBO J 2021; 40:e107192. [PMID: 33934370 PMCID: PMC8204859 DOI: 10.15252/embj.2020107192] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/26/2022] Open
Abstract
The lymphatic system is composed of a hierarchical network of fluid absorbing lymphatic capillaries and transporting collecting vessels. Despite distinct functions and morphologies, molecular mechanisms that regulate the identity of the different vessel types are poorly understood. Through transcriptional analysis of murine dermal lymphatic endothelial cells (LECs), we identified Foxp2, a member of the FOXP family of transcription factors implicated in speech development, as a collecting vessel signature gene. FOXP2 expression was induced after initiation of lymph flow in vivo and upon shear stress on primary LECs in vitro. Loss of FOXC2, the major flow-responsive transcriptional regulator of lymphatic valve formation, abolished FOXP2 induction in vitro and in vivo. Genetic deletion of Foxp2 in mice using the endothelial-specific Tie2-Cre or the tamoxifen-inducible LEC-specific Prox1-CreERT2 line resulted in enlarged collecting vessels and defective valves characterized by loss of NFATc1 activity. Our results identify FOXP2 as a new flow-induced transcriptional regulator of collecting lymphatic vessel morphogenesis and highlight the existence of unique transcription factor codes in the establishment of vessel-type-specific endothelial cell identities.
Collapse
Affiliation(s)
| | - Maria H Ulvmar
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Alejandra González-Loyola
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Ioannis Kritikos
- Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
| | - Ying Sun
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
| | - Tatiana V Petrova
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
9
|
Benazzo A, Cho A, Nechay A, Schwarz S, Frommlet F, Wekerle T, Hoetzenecker K, Jaksch P. Combined low-dose everolimus and low-dose tacrolimus after Alemtuzumab induction therapy: a randomized prospective trial in lung transplantation. Trials 2021; 22:6. [PMID: 33397442 PMCID: PMC7783986 DOI: 10.1186/s13063-020-04843-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/23/2020] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Long-term outcomes of lung transplantation are severely affected by comorbidities and development of chronic rejection. Among the comorbidities, kidney insufficiency is one of the most frequent and it is mainly caused by the cumulative effect of calcineurin inhibitors (CNIs). Currently, the most used immunosuppression protocols worldwide include induction therapy and a triple-drug maintenance immunosuppression, with one calcineurin inhibitor, one anti-proliferative drug, and steroids. Our center has pioneered the use of alemtuzumab as induction therapy, showing promising results in terms of short- and long-term outcomes. The use of alemtuzumab followed by a low-dose double drug maintenance immunosuppression, in fact, led to better kidney function along with excellent results in terms of acute rejection, chronic lung allograft dysfunction, and survival (Benazzo et al., PLoS One 14(1):e0210443, 2019). The hypothesis driving the proposed clinical trial is that de novo introduction of low-dose everolimus early after transplantation could further improve kidney function via a further reduction of tacrolimus. Based on evidences from kidney transplantation, moreover, alemtuzumab induction therapy followed by a low-dose everolimus and low-dose tacrolimus may have a permissive action on regulatory immune cells thus stimulating allograft acceptance. METHODS A randomized prospective clinical trial has been set up to answer the research hypothesis. One hundred ten patients will be randomized in two groups. Treatment group will receive the new maintenance immunosuppression protocol based on low-dose tacrolimus and low-dose everolimus and the control group will receive our standard immunosuppression protocol. Both groups will receive alemtuzumab induction therapy. The primary endpoint of the study is to analyze the effect of the new low-dose immunosuppression protocol on kidney function in terms of eGFR change. The study will have a duration of 24 months from the time of randomization. Immunomodulatory status of the patients will be assessed with flow cytometry and gene expression analysis. DISCUSSION For the first time in the field of lung transplantation, this trial proposes the combined use of significantly reduced tacrolimus and everolimus after alemtuzumab induction. The new protocol may have a twofold advantage: (1) further reduction of nephrotoxic tacrolimus and (2) permissive influence on regulatory cells development with further reduction of rejection episodes. TRIAL REGISTRATION EUDRACT Nr 2018-001680-24. Registered on 15 May 2018.
Collapse
Affiliation(s)
| | - Ara Cho
- Medizinische Universitat Wien, Vienna, Austria
| | - Anna Nechay
- Medizinische Universitat Wien, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
10
|
Meško M, Lebar T, Dekleva P, Jerala R, Benčina M. Engineering and Rewiring of a Calcium-Dependent Signaling Pathway. ACS Synth Biol 2020; 9:2055-2065. [PMID: 32643923 PMCID: PMC7467823 DOI: 10.1021/acssynbio.0c00133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
![]()
An important feature of synthetic
biological circuits is their
response to physicochemical signals, which enables the external control
of cellular processes. Calcium-dependent regulation is an attractive
approach for achieving such control, as diverse stimuli induce calcium
influx by activating membrane channel receptors. Most calcium-dependent
gene circuits use the endogenous nuclear factor of activated T-cells
(NFAT) signaling pathway. Here, we employed engineered NFAT transcription
factors to induce the potent and robust activation of exogenous gene
expression in HEK293T cells. Furthermore, we designed a calcium-dependent
transcription factor that does not interfere with NFAT-regulated promoters
and potently activates transcription in several mammalian cell types.
Additionally, we demonstrate that coupling the circuit to a calcium-selective
ion channel resulted in capsaicin- and temperature-controlled gene
expression. This engineered calcium-dependent signaling pathway enables
tightly controlled regulation of gene expression through different
stimuli in mammalian cells and is versatile, adaptable, and useful
for a wide range of therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Maja Meško
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
- Interfaculty Doctoral Study of Biomedicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Tina Lebar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Petra Dekleva
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, SI-1000 Ljubljana, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
11
|
Lee JU, Kim LK, Choi JM. Revisiting the Concept of Targeting NFAT to Control T Cell Immunity and Autoimmune Diseases. Front Immunol 2018; 9:2747. [PMID: 30538703 PMCID: PMC6277705 DOI: 10.3389/fimmu.2018.02747] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/08/2018] [Indexed: 01/15/2023] Open
Abstract
The nuclear factor of activated T cells (NFAT) family of transcription factors, which includes NFAT1, NFAT2, and NFAT4, are well-known to play important roles in T cell activation. Most of NFAT proteins are controlled by calcium influx upon T cell receptor and costimulatory signaling results increase of IL-2 and IL-2 receptor. NFAT3 however is not shown to be expressed in T cells and NFAT5 has not much highlighted in T cell functions yet. Recent studies demonstrate that the NFAT family proteins involve in function of lineage-specific transcription factors during differentiation of T helper 1 (Th1), Th2, Th17, regulatory T (Treg), and follicular helper T cells (Tfh). They have been studied to make physical interaction with the other transcription factors like GATA3 or Foxp3 and they also regulate Th cell signature gene expressions by direct binding on promotor region of target genes. From last decades, NFAT functions in T cells have been targeted to develop immune modulatory drugs for controlling T cell immunity in autoimmune diseases like cyclosporine A, FK506, etc. Due to their undesirable side defects, only limited application is available in human diseases. This review focuses on the recent advances in development of NFAT targeting drug as well as our understanding of each NFAT family protein in T cell biology. We also discuss updated detail molecular mechanism of NFAT functions in T cells, which would lead us to suggest an idea for developing specific NFAT inhibitors as a therapeutic drug for autoimmune diseases.
Collapse
Affiliation(s)
- Jae-Ung Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Li-Kyung Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| |
Collapse
|
12
|
Wallin EF, Hill DL, Linterman MA, Wood KJ. The Calcineurin Inhibitor Tacrolimus Specifically Suppresses Human T Follicular Helper Cells. Front Immunol 2018; 9:1184. [PMID: 29904381 PMCID: PMC5990622 DOI: 10.3389/fimmu.2018.01184] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023] Open
Abstract
Background T follicular helper (Tfh) cells are key players in the production of antibody-producing B cells via the germinal center reaction. Therapeutic strategies targeting Tfh cells are important where antibody formation is implicated in disease, such as transplant rejection and autoimmune diseases. We investigated the impact of the immunosuppressive agent tacrolimus on human Tfh cell differentiation and function in transplant recipients. Methods Paired blood and lymph node (LN) samples were obtained from 61 transplant recipients immediately prior to organ implantation. Living-donor recipients received a week of tacrolimus prior to kidney transplantation. Deceased-donor recipients served as controls, as tacrolimus was not administered until after the transplant operation. Flow cytometry was used to compare LN and circulating cell subsets. Results The calcineurin inhibitor (CNIs) tacrolimus specifically suppresses both LN Tfh cells and circulating Tfh cells, but not their regulatory counterparts or other CD4 T cell subsets. Conclusion Our findings suggest that CNIs may have a more important role in the prevention of antibody formation than previously understood and, therefore, have potential for antibody-associated conditions in which aberrant Tfh function has been implicated in disease.
Collapse
Affiliation(s)
- Elizabeth F Wallin
- Transplant Research Immunology Group, Nuffield Department Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Danika L Hill
- Lymphocyte Signalling ISP, Babraham Institute, Cambridge, United Kingdom.,Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | | | - Kathryn J Wood
- Transplant Research Immunology Group, Nuffield Department Surgical Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Agüera-González S, Burton OT, Vázquez-Chávez E, Cuche C, Herit F, Bouchet J, Lasserre R, Del Río-Iñiguez I, Di Bartolo V, Alcover A. Adenomatous Polyposis Coli Defines Treg Differentiation and Anti-inflammatory Function through Microtubule-Mediated NFAT Localization. Cell Rep 2018; 21:181-194. [PMID: 28978472 DOI: 10.1016/j.celrep.2017.09.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/04/2017] [Accepted: 09/05/2017] [Indexed: 10/18/2022] Open
Abstract
Adenomatous polyposis coli (APC) is a polarity regulator and tumor suppressor associated with familial adenomatous polyposis and colorectal cancer development. Although extensively studied in epithelial transformation, the effect of APC on T lymphocyte activation remains poorly defined. We found that APC ensures T cell receptor-triggered activation through Nuclear Factor of Activated T cells (NFAT), since APC is necessary for NFAT's nuclear localization in a microtubule-dependent fashion and for NFAT-driven transcription leading to cytokine gene expression. Interestingly, NFAT forms clusters juxtaposed with microtubules. Ultimately, mouse Apc deficiency reduces the presence of NFAT in the nucleus of intestinal regulatory T cells (Tregs) and impairs Treg differentiation and the acquisition of a suppressive phenotype, which is characterized by the production of the anti-inflammatory cytokine IL-10. These findings suggest a dual role for APC mutations in colorectal cancer development, where mutations drive the initiation of epithelial neoplasms and also reduce Treg-mediated suppression of the detrimental inflammation that enhances cancer growth.
Collapse
Affiliation(s)
- Sonia Agüera-González
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France.
| | - Oliver T Burton
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Vázquez-Chávez
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Céline Cuche
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Floriane Herit
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Jérôme Bouchet
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Rémi Lasserre
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Iratxe Del Río-Iñiguez
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Vincenzo Di Bartolo
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Andrés Alcover
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France.
| |
Collapse
|
14
|
Marangoni F, Zhang R, Mani V, Thelen M, Ali Akbar NJ, Warner RD, Äijö T, Zappulli V, Martinez GJ, Turka LA, Mempel TR. Tumor Tolerance-Promoting Function of Regulatory T Cells Is Optimized by CD28, but Strictly Dependent on Calcineurin. THE JOURNAL OF IMMUNOLOGY 2018; 200:3647-3661. [PMID: 29661826 DOI: 10.4049/jimmunol.1701220] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/13/2018] [Indexed: 01/07/2023]
Abstract
Regulatory T cells (Treg) restrain immune responses against malignant tumors, but their global depletion in cancer patients will likely be limited by systemic autoimmune toxicity. Instead, approaches to "tune" their activities may allow for preferential targeting of tumor-reactive Treg. Although Ag recognition regulates Treg function, the roles of individual TCR-dependent signaling pathways in enabling Treg to promote tumor tolerance are not well characterized. In this study, we examined in mouse tumor models the role of calcineurin, a key mediator of TCR signaling, and the role of the costimulatory receptor CD28 in the differentiation of resting central Treg into effector Treg endowed with tumor tropism. We find that calcineurin, although largely dispensable for suppressive activity in vitro, is essential for upregulation of ICOS and CTLA-4 in Treg, as well as for expression of chemokine receptors driving their accumulation in tumors. In contrast, CD28 is not critical, but optimizes the formation of tumor-homing Treg and their fitness in tumor tissue. Accordingly, although deletion of either CnB or CD28 strongly impairs Treg-mediated tumor tolerance, lack of CnB has an even more pronounced impact than lack of CD28. Hence, our studies reveal distinct roles for what has classically been defined as signal 1 and signal 2 of conventional T cell activation in the context of Treg-mediated tumor tolerance.
Collapse
Affiliation(s)
- Francesco Marangoni
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114; .,Harvard Medical School, Boston, MA 02115
| | - Ruan Zhang
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02114
| | - Vinidhra Mani
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114.,Harvard Medical School, Boston, MA 02115
| | - Martin Thelen
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Noor J Ali Akbar
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Ross D Warner
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Tarmo Äijö
- Center for Computational Biology, Flatiron Institute, New York, NY 10010
| | - Valentina Zappulli
- Department of Comparative Biomedicine and Food Science, University of Padua, 35020 Legnaro, Padua, Italy; and
| | - Gustavo J Martinez
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Laurence A Turka
- Harvard Medical School, Boston, MA 02115.,Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02114
| | - Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114; .,Harvard Medical School, Boston, MA 02115
| |
Collapse
|
15
|
Ram BM, Dolpady J, Kulkarni R, Usha R, Bhoria U, Poli UR, Islam M, Trehanpati N, Ramakrishna G. Human papillomavirus (HPV) oncoprotein E6 facilitates Calcineurin-Nuclear factor for activated T cells 2 (NFAT2) signaling to promote cellular proliferation in cervical cell carcinoma. Exp Cell Res 2018; 362:132-141. [DOI: 10.1016/j.yexcr.2017.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022]
|
16
|
Whitehouse G, Gray E, Mastoridis S, Merritt E, Kodela E, Yang JHM, Danger R, Mairal M, Christakoudi S, Lozano JJ, Macdougall IC, Tree TIM, Sanchez-Fueyo A, Martinez-Llordella M. IL-2 therapy restores regulatory T-cell dysfunction induced by calcineurin inhibitors. Proc Natl Acad Sci U S A 2017; 114:7083-7088. [PMID: 28584086 PMCID: PMC5502598 DOI: 10.1073/pnas.1620835114] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
CD4+CD25+FOXP3+ Tregs constitute a heterogeneous lymphocyte subpopulation essential for curtailing effector T cells and establishing peripheral tolerance. Calcineurin inhibitors (CNIs) are among the most effective agents in controlling effector T-cell responses in humans. However, CNIs also reduce the size of the Treg pool. The functional consequences of this negative effect and the mechanisms responsible remain to be elucidated. We report here that CNIs compromise the overall Treg immunoregulatory capacity to a greater extent than would be predicted by the reduction in the size of the Treg compartment, given that they selectively promote the apoptosis of the resting and activated Treg subsets that are known to display the most powerful suppressive function. These effects are caused by reduced access to IL-2, because Tregs remain capable of translocating NFAT even in the presence of high CNI levels. Exogenous IL-2 restores the phenotypic changes and overall gene-expression effects exerted by CNIs and can even promote Treg expansion by enhancing antiapoptotic Bcl-2 expression. In a skin transplant model, the addition of IL-2 synergizes with CNIs treatment, promoting intragraft accumulation of Tregs and prolonged allograft survival. Hence, the combination of IL-2 and CNIs constitutes an optimal immunomodulatory regimen that enhances the pool of suppressive Treg subsets while effectively controlling cytopathic T cells.
Collapse
Affiliation(s)
- Gavin Whitehouse
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| | - Elizabeth Gray
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| | - Sotiris Mastoridis
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| | - Elliot Merritt
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| | - Elisavet Kodela
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| | - Jennie H M Yang
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, United Kingdom
| | - Richard Danger
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| | - Marta Mairal
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| | - Sofia Christakoudi
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| | - Juan J Lozano
- Bioinformatics Platform, Biomedical Research Networking Center in Hepatic and Digestive Diseases, Barcelona 08036, Spain
| | - Iain C Macdougall
- Department of Renal Medicine, King's College Hospital, London SE5 9RS, United Kingdom
| | - Timothy I M Tree
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, United Kingdom
| | - Alberto Sanchez-Fueyo
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| | - Marc Martinez-Llordella
- Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London SE5 9RS, United Kingdom;
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, United Kingdom
| |
Collapse
|
17
|
Li J, Huang L, Wang S, Yao Y, Zhang Z. Astragaloside IV attenuates inflammatory reaction via activating immune function of regulatory T-cells inhibited by HMGB1 in mice. PHARMACEUTICAL BIOLOGY 2016; 54:3217-3225. [PMID: 27564970 DOI: 10.1080/13880209.2016.1216133] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/03/2016] [Accepted: 07/19/2016] [Indexed: 05/19/2023]
Abstract
CONTEXT High-mobility group box 1 (HMGB1) protein is a highly abundant protein that can promote the pathogenesis of inflammatory. Some experiments have demonstrated a vital role for HMGB1 to modulate the immune function of regulatory T-cells (Tregs). Astragaloside IV (AST IV), an extract from Astragalus membranaceus Moench (Leguminosae), has been shown to exert potent cardioprotective and anti-inflammatory effects. It is still unclear whether AST IV has a latent effect on the proinflammatory ability of HMGB1 with subsequent activation of Tregs in vivo. OBJECTIVE This research explores the antagonism of different doses of AST IV on the immunologic function of Tregs mediated by HMGB1. MATERIALS AND METHODS Mouse models (BALB/c) were constructed by which normal saline or AST IV was administered i.p. at 2, 4 and 6 days after the administration i.p. of 20 μg recombinate HMGB1. Spleen was used to procure Treg and CD4 + CD25- T-cells which were co-cultured with Treg. Cell phenotypes of Tregs(Foxp3) were examined, and the cytokine levels in supernatants and the proliferation of T-cells were assayed. Gene expression was measured by RT-PCR. RESULTS (1) The expression levels of Foxp3 in Treg on post-stimulus days (PSD) 1-7 were significantly decreased in the HMGB1 group in comparison to those in the control group mice (p < 0.01). The Foxp3 expression was markedly increased in a dose-dependent manner in the AST group as compared with those in the HMGB1 group (p < 0.0 1-0.05). The same results were found in the contents of cytokines (IL-10 and TGF-β) released into supernatants by Treg. (2) When CD4 + CD25- T-cells were co-cultured with Treg stimulated by HMGB1, the cell proliferation and the levels of cytokines (IL-2 and IFN-γ) in supernatant were markedly increased as compared with those in the HMGB1 group. The level of IL-4 was markedly decreased as compared with that in the HMGB1 group. The same results were found when CD4 + CD25- T-cells were co-cultured with Treg in the NS group. Compared with those in the NS group, the contrary results were shown in a dose-dependent manner in the AST group. DISCUSSION AND CONCLUSION These results showed that AST IV has a therapeutic effect on inflammation promoted by HMGB1, and it should be studied as a new drug for the treatment of sepsis.
Collapse
Affiliation(s)
- Jinfeng Li
- a Department of Obstetrics and Gynecology , Beijing Chao-Yang Hospital, Capital Medical University , Beijing , China
| | - Lifeng Huang
- b Department of Surgical Intensive Care Unit , Beijing Chao-Yang Hospital, Capital Medical University , Beijing , China
| | - Shuzhen Wang
- a Department of Obstetrics and Gynecology , Beijing Chao-Yang Hospital, Capital Medical University , Beijing , China
| | - Yongming Yao
- c Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital , Beijing , China
| | - Zhenyu Zhang
- a Department of Obstetrics and Gynecology , Beijing Chao-Yang Hospital, Capital Medical University , Beijing , China
| |
Collapse
|
18
|
Schmiedeberg K, Krause H, Röhl FW, Hartig R, Jorch G, Brunner-Weinzierl MC. T Cells of Infants Are Mature, but Hyporeactive Due to Limited Ca2+ Influx. PLoS One 2016; 11:e0166633. [PMID: 27893767 PMCID: PMC5125607 DOI: 10.1371/journal.pone.0166633] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/01/2016] [Indexed: 12/12/2022] Open
Abstract
CD4 T cells in human infants and adults differ in the initiation and strength of their responses. The molecular basis for these differences is not yet understood. To address this the principle key molecular events of TCR- and CD28-induced signaling in naive CD4 T cells, such as Ca2+ influx, NFAT expression, phosphorylation and translocation into the nucleus, ERK activation and IL-2 response, were analyzed over at least the first 3 years of life. We report dramatically reduced IL-2 and TNFα responses in naive CD31+ T cells during infancy. Looking at the obligatory Ca2+ influx required to induce T cell activation and proliferation, we demonstrate characteristic patterns of impairment for each stage of infancy that are partly due to the differential usage of Ca2+ stores. Consistent with those findings, translocation of NFATc2 is limited, but still dependent on Ca2+ influx as demonstrated by sensitivity to cyclosporin A (CsA) treatment. Thus weak Ca2+ influx functions as a catalyst for the implementation of restricted IL-2 response in T cells during infancy. Our studies also define limited mobilization of Ca2+ ions as a characteristic property of T cells during infancy. This work adds to our understanding of infants’ poor T cell responsiveness against pathogens.
Collapse
Affiliation(s)
- Kristin Schmiedeberg
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Hardy Krause
- Clinic of Pediatric Surgery University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Friedrich-Wilhelm Röhl
- Institute of Biometry and Medical Informatics University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Roland Hartig
- Institute for Molecular and Clinical Immunology, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Gerhard Jorch
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Monika C. Brunner-Weinzierl
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
- * E-mail:
| |
Collapse
|
19
|
Abstract
Immunosuppression strategies that selectively inhibit effector T cells while preserving and even enhancing CD4FOXP3 regulatory T cells (Treg) permit immune self-regulation and may allow minimization of immunosuppression and associated toxicities. Many immunosuppressive drugs were developed before the identity and function of Treg were appreciated. A good understanding of the interactions between Treg and immunosuppressive agents will be valuable to the effective design of more tolerable immunosuppression regimens. This review will discuss preclinical and clinical evidence regarding the influence of current and emerging immunosuppressive drugs on Treg homeostasis, stability, and function as a guideline for the selection and development of Treg-friendly immunosuppressive regimens.
Collapse
Affiliation(s)
- Akiko Furukawa
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Steven A Wisel
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
20
|
de Almeida Nagata DE, Ting HA, Cavassani KA, Schaller MA, Mukherjee S, Ptaschinski C, Kunkel SL, Lukacs NW. Epigenetic control of Foxp3 by SMYD3 H3K4 histone methyltransferase controls iTreg development and regulates pathogenic T-cell responses during pulmonary viral infection. Mucosal Immunol 2015; 8:1131-43. [PMID: 25669152 PMCID: PMC4532649 DOI: 10.1038/mi.2015.4] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/02/2015] [Indexed: 02/04/2023]
Abstract
The generation of regulatory T (Treg) cells is driven by Foxp3 and is responsible for dampening inflammation and reducing autoimmunity. In this study, the epigenetic regulation of inducible Treg (iTreg) cells was examined and an H3K4 histone methyltransferase, SMYD3 (SET and MYND Domain 3), which regulates the expression of Foxp3 by a TGFβ1/Smad3 (transforming growth factor-β1/Smad3)-dependent mechanism, was identified. Using chromatin immunoprecipitation assays, SMYD3 depletion led to a reduction in H3K4me3 in the promoter region and CNS1 (conserved noncoding DNA sequence) of the foxp3 locus. SMYD3 abrogation affected iTreg cell formation while allowing dysregulated interleukin-17 production. In a mouse model of respiratory syncytial virus (RSV) infection, a model in which iTreg cells have a critical role in regulating lung pathogenesis, SMYD3(-/-) mice demonstrated exacerbation of RSV-induced disease related to enhanced proinflammatory responses and worsened pathogenesis within the lung. Our data highlight a novel activation role for the TGFβ-inducible SMYD3 in regulating iTreg cell formation leading to increased severity of virus-related disease.
Collapse
Affiliation(s)
| | - Hung-An Ting
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Karen A. Cavassani
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Matthew A. Schaller
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Sumanta Mukherjee
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Steven L. Kunkel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Nicholas W. Lukacs
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|
21
|
Pinto MCX, Kihara AH, Goulart VAM, Tonelli FMP, Gomes KN, Ulrich H, Resende RR. Calcium signaling and cell proliferation. Cell Signal 2015; 27:2139-49. [PMID: 26275497 DOI: 10.1016/j.cellsig.2015.08.006] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/04/2015] [Accepted: 08/10/2015] [Indexed: 12/17/2022]
Abstract
Cell proliferation is orchestrated through diverse proteins related to calcium (Ca(2+)) signaling inside the cell. Cellular Ca(2+) influx that occurs first by various mechanisms at the plasma membrane, is then followed by absorption of Ca(2+) ions by mitochondria and endoplasmic reticulum, and, finally, there is a connection of calcium stores to the nucleus. Experimental evidence indicates that the fluctuation of Ca(2+) from the endoplasmic reticulum provides a pivotal and physiological role for cell proliferation. Ca(2+) depletion in the endoplasmatic reticulum triggers Ca(2+) influx across the plasma membrane in an phenomenon called store-operated calcium entries (SOCEs). SOCE is activated through a complex interplay between a Ca(2+) sensor, denominated STIM, localized in the endoplasmic reticulum and a Ca(2+) channel at the cell membrane, denominated Orai. The interplay between STIM and Orai proteins with cell membrane receptors and their role in cell proliferation is discussed in this review.
Collapse
Affiliation(s)
- Mauro Cunha Xavier Pinto
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Presyes 748, 05508-000 São Paulo, SP, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Alexandre Hiroaki Kihara
- Universidade Federal do ABC, Centro de Matemática, Computação e Cognição, Rua Arcturus (Jd Antares), 09606-070, São Bernardo do Campo, SP, Brazil
| | - Vânia A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Fernanda M P Tonelli
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Katia N Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Presyes 748, 05508-000 São Paulo, SP, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
22
|
Fanigliulo D, Lazzerini PE, Capecchi PL, Ulivieri C, Baldari CT, Laghi-Pasini F. Clinically-relevant cyclosporin and rapamycin concentrations enhance regulatory T cell function to a similar extent but with different mechanisms: an in-vitro study in healthy humans. Int Immunopharmacol 2014; 24:276-284. [PMID: 25536542 DOI: 10.1016/j.intimp.2014.12.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 12/31/2022]
Abstract
Evidence indicates that regulatory T cells (Tregs) are profoundly involved in promoting allograft tolerance after organ transplantation. Since a successful transplantation currently still requires a long-term immunosuppressive treatment, clarifying the specific impact of these drugs on Tregs may be of high clinical relevance. Conflicting results arise from the literature, particularly as concerns cyclosporine (CsA). The specific aim of this work was to evaluate in-vitro the direct effects of clinically-relevant drug concentrations of three widely used immunosuppressive drugs, i.e. CsA, rapamycin (RAPA) and mycophenolic acid (MPA), on Treg activity, number and forkhead/winged helix transcription factor (FoxP3) expression in humans. Tregs (CD4(+)CD25(+)) isolated from healthy donors were cultured in the presence of different concentrations of CsA, RAPA or MPA. The suppressive activity of Tregs was evaluated in mixed lymphocyte reactions with CD4(+)CD25(-) T cells. Phenotype analysis and FoxP3 expression were assessed by flow cytometry. Clinically-relevant CsA and RAPA concentrations significantly enhanced to a similar extent the suppressive activity of Tregs. Although this effect was associated with an increase in Treg number as well as in FoxP3 expression with both drugs, the driving mechanism seemed to be primarily quantitative (i.e. increase of the cell number) for RAPA, whereas mainly qualitative (i.e. increase in FoxP3 levels) for CsA, respectively. Conversely, MPA did not show any effect on Treg function and number. These findings suggest that both RAPA and CsA may be beneficial in promoting Treg-dependent allograft tolerance after organ transplantation.
Collapse
Affiliation(s)
- Daniela Fanigliulo
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, Siena, Italy
| | - Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, Policlinico Le Scotte, University of Siena, Viale Bracci, Siena, Italy.
| | - Pier Leopoldo Capecchi
- Department of Medical Sciences, Surgery and Neurosciences, Policlinico Le Scotte, University of Siena, Viale Bracci, Siena, Italy
| | - Cristina Ulivieri
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, Siena, Italy
| | | | - Franco Laghi-Pasini
- Department of Medical Sciences, Surgery and Neurosciences, Policlinico Le Scotte, University of Siena, Viale Bracci, Siena, Italy
| |
Collapse
|
23
|
Qin JJ, Nag S, Wang W, Zhou J, Zhang WD, Wang H, Zhang R. NFAT as cancer target: mission possible? Biochim Biophys Acta Rev Cancer 2014; 1846:297-311. [PMID: 25072963 DOI: 10.1016/j.bbcan.2014.07.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 07/17/2014] [Accepted: 07/22/2014] [Indexed: 12/30/2022]
Abstract
The NFAT signaling pathway regulates various aspects of cellular functions; NFAT acts as a calcium sensor, integrating calcium signaling with other pathways involved in development and growth, immune response, and inflammatory response. The NFAT family of transcription factors regulates diverse cellular functions such as cell survival, proliferation, migration, invasion, and angiogenesis. The NFAT isoforms are constitutively activated and overexpressed in several cancer types wherein they transactivate downstream targets that play important roles in cancer development and progression. Though the NFAT family has been conclusively proved to be pivotal in cancer progression, the different isoforms play distinct roles in different cellular contexts. In this review, our discussion is focused on the mechanisms that drive the activation of various NFAT isoforms in cancer. Additionally, we analyze the potential of NFAT as a valid target for cancer prevention and therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Subhasree Nag
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Jianwei Zhou
- Department of Molecular Cell Biology and Toxicology, Cancer Center, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Wei-Dong Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Hui Wang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, PR China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing 100021, PR China
| | - Ruiwen Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
24
|
Li W, Kong LB, Li JT, Guo ZY, Xue Q, Yang T, Meng YL, Jin BQ, Wen WH, Yang AG. MiR-568 inhibits the activation and function of CD4⁺ T cells and Treg cells by targeting NFAT5. Int Immunol 2013; 26:269-81. [PMID: 24355664 DOI: 10.1093/intimm/dxt065] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
CD4(+) T cells play critical roles in orchestrating adaptive immune responses. Their activation and proliferation are critical steps that occur before they execute their biological functions. Despite the important role of this process, the underlying molecular events are not fully understood. MicroRNAs (miRNAs) have been shown to play important roles in lymphocyte development and function. However, the miRNAs that regulate T-cell differentiation, activation and proliferation are still largely unknown. In our previous study, using a miRNA array, we found that several miRNAs (including miR-202, 33b, 181c, 568 and 576) are differentially expressed between resting and activated CD4(+) T cells. In this study, we focused on the function of miR-568 during CD4(+) T-cell activation. We showed that the expression level of miR-568 decreased during the activation of T cells, including Jurkat cells and human peripheral blood CD4(+) T cells. When Jurkat or human peripheral blood CD4(+) T cells were transfected with miR-568 mimics, cell activation was significantly inhibited, as shown by the inhibited expression of activation markers such as CD25, CD69 and CD154; decreased IL-2 production; and inhibited cell proliferation. Using software predictions and confirmatory experiments, we demonstrated that nuclear factor of activated T cells 5 (NFAT5) is a target of miR-568. Treg cells are an important CD4(+) T-cell subpopulation, so we also evaluated the function of miR-568 in Treg-cell activation and differentiation. We showed that the miR-568 level decreased, while the NFAT5 protein level increased during CD4(+)CD25(+) Treg-cell activation, and the transfection of miR-568 mimics inhibited the NFAT5 expression, inhibited the production of both TGF-β and IL-10 and also inhibited the proliferation of Treg cells. Our further study showed that over-expression of miR-568 can inhibit Treg-cell differentiation and can inhibit the suppressive effect of these cells on effector cells. In addition, inhibition of NFAT5 by siRNA-mediated knockdown can inhibit the activation and differentiation of Treg cells. These findings reveal that miR-568 can inhibit the activation and function of both CD4(+) T cells and Treg cells by targeting NFAT5. Since miR-568 plays an important role in both CD4(+) T cells and Treg cells, these findings may provide leads for the development of novel treatments for human inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Thymic regulatory T cell development: role of signalling pathways and transcription factors. Clin Dev Immunol 2013; 2013:617595. [PMID: 24187564 PMCID: PMC3803129 DOI: 10.1155/2013/617595] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/21/2013] [Indexed: 01/07/2023]
Abstract
Regulatory T cells (Tregs) are a subset of CD4 T cells that are key mediators of immune tolerance. Most Tregs develop in the thymus. In this review we summarise recent findings on the role of diverse signalling pathways and downstream transcription factors in thymic Treg development.
Collapse
|
26
|
Tantin D, Voth WP, Shakya A. Efficient chromatin immunoprecipitation using limiting amounts of biomass. J Vis Exp 2013:e50064. [PMID: 23665589 DOI: 10.3791/50064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chromatin immunoprecipitation (ChIP) is a widely-used method for determining the interactions of different proteins with DNA in chromatin of living cells. Examples include sequence-specific DNA binding transcription factors, histones and their different modification states, enzymes such as RNA polymerases and ancillary factors, and DNA repair components. Despite its ubiquity, there is a lack of up-to-date, detailed methodologies for both bench preparation of material and for accurate analysis allowing quantitative metrics of interaction. Due to this lack of information, and also because, like any immunoprecipitation, conditions must be re-optimized for new sets of experimental conditions, the ChIP assay is susceptible to inaccurate or poorly quantitative results. Our protocol is ultimately derived from seminal work on transcription factor:DNA interactions(1,2) , but incorporates a number of improvements to sensitivity and reproducibility for difficult-to-obtain cell types. The protocol has been used successfully(3,4) , both using qPCR to quantify DNA enrichment, or using a semi-quantitative variant of the below protocol. This quantitative analysis of PCR-amplified material is performed computationally, and represents a limiting factor in the assay. Important controls and other considerations include the use of an isotype-matched antibody, as well as evaluation of a control region of genomic DNA, such as an intergenic region predicted not to be bound by the protein under study (or anticipated not to show changes under the experimental conditions). In addition, a standard curve of input material for every ChIP sample is used to derive absolute levels of enrichment in the experimental material. Use of standard curves helps to take into account differences between primer sets, regardless of how carefully they are designed, and also efficiency differences throughout the range of template concentrations for a single primer set. Our protocol is different from others that are available(5-8) in that we extensively cover the later, analysis phase.
Collapse
Affiliation(s)
- Dean Tantin
- Department of Pathology, University of Utah School of Medicine
| | | | | |
Collapse
|
27
|
Chaiyachati BH, Jani A, Wan Y, Huang H, Flavell R, Chi T. BRG1-mediated immune tolerance: facilitation of Treg activation and partial independence of chromatin remodelling. EMBO J 2013; 32:395-408. [PMID: 23321680 DOI: 10.1038/emboj.2012.350] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 12/05/2012] [Indexed: 02/02/2023] Open
Abstract
Treg activation in response to environmental cues is necessary for regulatory T cells (Tregs) to suppress inflammation, but little is known about the transcription mechanisms controlling Treg activation. We report that despite the known proinflammatory role of the chromatin-remodelling factor BRG1 in CD4 cells, deleting Brg1 in all αβ T cell lineages led to fatal inflammation, which reflected essential roles of BRG1 in Tregs. Brg1 deletion impaired Treg activation, concomitant with the onset of the inflammation. Remarkably, as the inflammation progressed, Tregs became increasingly activated, but the activation levels could not catch up with the severity of inflammation. In vitro assays indicate that BRG1 regulates a subset of TCR target genes including multiple chemokine receptor genes. Finally, using a method that can create littermates bearing either a tissue-specific point mutation or deletion, we found the BRG1 ATPase activity partially dispensable for BRG1 function. Collectively, these data suggest that BRG1 acts in part via remodelling-independent functions to sensitize Tregs to inflammatory cues, thus allowing Tregs to promptly and effectively suppress autoimmunity.
Collapse
Affiliation(s)
- Barbara H Chaiyachati
- Department of Immunobiology, Yale University Medical School, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
28
|
Scheel T, Abajyan A, Baumgrass R. Comment on "constitutive nuclear localization of NFAT in Foxp3(+) regulatory T cells independent of calcineurin activity". THE JOURNAL OF IMMUNOLOGY 2012; 189:4703; author reply 4703-4. [PMID: 23125425 DOI: 10.4049/jimmunol.1290068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
29
|
Heiber JF, Geiger TL. Context and location dependence of adaptive Foxp3(+) regulatory T cell formation during immunopathological conditions. Cell Immunol 2012; 279:60-5. [PMID: 23089195 DOI: 10.1016/j.cellimm.2012.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/29/2012] [Accepted: 09/12/2012] [Indexed: 02/07/2023]
Abstract
Circulating Foxp3(+) regulatory T cells (Treg) may arise in the thymus (natural Treg, nTreg) or through the adaptive upregulation of Foxp3 after T cell activation (induced Treg, iTreg). In this brief review, we explore evidence for the formation and function of iTreg during pathologic conditions. Determining the ontogeny and function of Treg populations has relied on the use of manipulated systems in which either iTreg or nTreg are absent, or lineage tracing of T cell clones through repertoire analyses. iTreg appear particularly important at mucosal interfaces. iTreg can also ameliorate tissue-specific autoimmunity and are a prominent source of tumor-infiltrating Treg in some models. However, under many conditions, including in CNS autoimmunity, diabetes, and some tumor systems, iTreg formation appears limited. The immunological contribution of iTreg is thus highly context dependent. Deciphering immune parameters responsible for iTreg formation and their role in modulating pathologic immune responses will be important.
Collapse
Affiliation(s)
- Joshua F Heiber
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
30
|
Dependence on nuclear factor of activated T-cells (NFAT) levels discriminates conventional T cells from Foxp3+ regulatory T cells. Proc Natl Acad Sci U S A 2012; 109:16258-63. [PMID: 22991461 DOI: 10.1073/pnas.1203870109] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Several lines of evidence suggest nuclear factor of activated T-cells (NFAT) to control regulatory T cells: thymus-derived naturally occurring regulatory T cells (nTreg) depend on calcium signals, the Foxp3 gene harbors several NFAT binding sites, and the Foxp3 (Fork head box P3) protein interacts with NFAT. Therefore, we investigated the impact of NFAT on Foxp3 expression. Indeed, the generation of peripherally induced Treg (iTreg) by TGF-β was highly dependent on NFAT expression because the ability of CD4(+) T cells to differentiate into iTreg diminished markedly with the number of NFAT family members missing. It can be concluded that the expression of Foxp3 in TGF-β-induced iTreg depends on the threshold value of NFAT rather than on an individual member present. This is specific for iTreg development, because frequency of nTreg remained unaltered in mice lacking NFAT1, NFAT2, or NFAT4 alone or in combination. Different from expectation, however, the function of both nTreg and iTreg was independent on robust NFAT levels, reflected by less nuclear NFAT in nTreg and iTreg. Accordingly, absence of one or two NFAT members did not alter suppressor activity in vitro or during colitis and transplantation in vivo. This scenario emphasizes an inhibition of high NFAT activity as treatment for autoimmune diseases and in transplantation, selectively targeting the proinflammatory conventional T cells, while keeping Treg functional.
Collapse
|
31
|
Huang LF, Yao YM, Li JF, Zhang SW, Li WX, Dong N, Yu Y, Sheng ZY. The effect of Astragaloside IV on immune function of regulatory T cell mediated by high mobility group box 1 protein in vitro. Fitoterapia 2012; 83:1514-22. [PMID: 22981502 DOI: 10.1016/j.fitote.2012.08.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/27/2012] [Accepted: 08/28/2012] [Indexed: 01/27/2023]
Abstract
High mobility group box 1 protein (HMGB1), a potent pro-inflammatory cytokine, contributes to the pathogenesis of diverse inflammatory and infectious disorders. Some studies have illustrated the potential effect of HMGB1 on regulatory T cells (Tregs). Astragaloside IV (AST IV) isolated from a Chinese herb, Astragalus mongholicus, is known to have a variety of immunomodulatory activities. However, it is not yet clear whether AST IV possesses potential regulatory effect on the pro-inflammatory ability of HMGB1 with subsequent activation of Tregs. This study was carried out to investigate the antagonistic effects of different doses of AST IV on the immune function of Tregs mediated by HMGB1 in vitro. Tregs isolated from the spleens of mice were co-cultured with HMGB1 and/or AST IV. Cell phenotypes of Tregs were analyzed, and the contents of various cytokines in the cell supernatants as a result of co-culture and the proliferation of CD4(+)CD25(-) T cells were determined. Results showed that HMGB1 stimulation resulted in significantly down-regulation of expressions of Tregs cell phenotypes. However, AST IV can rival the suppressing effect of HMGB1 on immune function of Tregs with a dose-dependent in vitro. These results indicate that AST IV has the potential therapeutic action on inflammation augmented by HMGB1.
Collapse
Affiliation(s)
- Li-feng Huang
- Department of Critical Care Medicine, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing 100050, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|