1
|
Horwitz JK, Bin S, Fairchild RL, Keslar KS, Yi Z, Zhang W, Pavlov VI, Li Y, Madsen JC, Cravedi P, Heeger PS. Linking erythropoietin to regulatory T-cell-dependent allograft survival through myeloid cells. JCI Insight 2022; 7:158856. [PMID: 35389892 PMCID: PMC9220923 DOI: 10.1172/jci.insight.158856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/06/2022] [Indexed: 12/03/2022] Open
Abstract
Erythropoietin (EPO) has multiple nonerythropoietic functions, including immune modulation, but EPO’s effects in transplantation remain incompletely understood. We tested the mechanisms linking EPO administration to prolongation of murine heterotopic heart transplantation using WT and conditional EPO receptor–knockout (EPOR-knockout) mice as recipients. In WT controls, peritransplant administration of EPO synergized with CTLA4-Ig to prolong allograft survival (P < 0.001), reduce frequencies of donor-reactive effector CD8+ T cells in the spleen (P < 0.001) and in the graft (P < 0.05), and increase frequencies and total numbers of donor-reactive Tregs (P < 0.01 for each) versus CTLA4-Ig alone. Studies performed in conditional EPOR-knockout recipients showed that each of these differences required EPOR expression in myeloid cells but not in T cells. Analysis of mRNA isolated from spleen monocytes showed that EPO/EPOR ligation upregulated macrophage-expressed, antiinflammatory, regulatory, and pro-efferocytosis genes and downregulated selected proinflammatory genes. Taken together, the data support the conclusion that EPO promotes Treg-dependent murine cardiac allograft survival by crucially altering the phenotype and function of macrophages. Coupled with our previous documentation that EPO promotes Treg expansion in humans, the data support the need for testing the addition of EPO to costimulatory blockade-containing immunosuppression regimens in an effort to prolong human transplant survival.
Collapse
Affiliation(s)
- Julian K Horwitz
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Sofia Bin
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Robert L Fairchild
- Department of Immunology, Cleveland Clinic, Cleveland, United States of America
| | - Karen S Keslar
- Department of Immunology, Cleveland Clinic, Cleveland, United States of America
| | - Zhengzi Yi
- Translational Transplant Research Center, Icahn School of medicine at Mount Sinai, New York, United States of America
| | - Weijia Zhang
- Translational Transplant Research Center, Icahn school of Medicine at Mount Sinai, New York, United States of America
| | - Vasile I Pavlov
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Yansui Li
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Joren C Madsen
- Department of Surgery, Massachusetts General Hospital, Boston, United States of America
| | - Paolo Cravedi
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Peter S Heeger
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| |
Collapse
|
2
|
Cristoferi I, Giacon TA, Boer K, van Baardwijk M, Neri F, Campisi M, Kimenai HJAN, Clahsen-van Groningen MC, Pavanello S, Furian L, Minnee RC. The applications of DNA methylation as a biomarker in kidney transplantation: a systematic review. Clin Epigenetics 2022; 14:20. [PMID: 35130936 PMCID: PMC8822833 DOI: 10.1186/s13148-022-01241-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/27/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Although kidney transplantation improves patient survival and quality of life, long-term results are hampered by both immune- and non-immune-mediated complications. Current biomarkers of post-transplant complications, such as allograft rejection, chronic renal allograft dysfunction, and cutaneous squamous cell carcinoma, have a suboptimal predictive value. DNA methylation is an epigenetic modification that directly affects gene expression and plays an important role in processes such as ischemia/reperfusion injury, fibrosis, and alloreactive immune response. Novel techniques can quickly assess the DNA methylation status of multiple loci in different cell types, allowing a deep and interesting study of cells' activity and function. Therefore, DNA methylation has the potential to become an important biomarker for prediction and monitoring in kidney transplantation. PURPOSE OF THE STUDY The aim of this study was to evaluate the role of DNA methylation as a potential biomarker of graft survival and complications development in kidney transplantation. MATERIAL AND METHODS: A systematic review of several databases has been conducted. The Newcastle-Ottawa scale and the Jadad scale have been used to assess the risk of bias for observational and randomized studies, respectively. RESULTS Twenty articles reporting on DNA methylation as a biomarker for kidney transplantation were included, all using DNA methylation for prediction and monitoring. DNA methylation pattern alterations in cells isolated from different tissues, such as kidney biopsies, urine, and blood, have been associated with ischemia-reperfusion injury and chronic renal allograft dysfunction. These alterations occurred in different and specific loci. DNA methylation status has also proved to be important for immune response modulation, having a crucial role in regulatory T cell definition and activity. Research also focused on a better understanding of the role of this epigenetic modification assessment for regulatory T cells isolation and expansion for future tolerance induction-oriented therapies. CONCLUSIONS Studies included in this review are heterogeneous in study design, biological samples, and outcome. More coordinated investigations are needed to affirm DNA methylation as a clinically relevant biomarker important for prevention, monitoring, and intervention.
Collapse
Affiliation(s)
- Iacopo Cristoferi
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands.
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands.
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands.
| | - Tommaso Antonio Giacon
- Kidney and Pancreas Transplantation Unit, Department of Surgical, Oncological and Gastroenterological Sciences, Padua University Hospital, Via Giustiniani 2, 35128, Padua, Italy
- Occupational Medicine, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padua University, Via Giustiniani 2, 35128, Padua, Italy
- Environmental and Respiratory Physiology Laboratory, Department of Biomedical Sciences, Padua University, Via Marzolo 3, 35131, Padua, Italy
- Institute of Anaesthesia and Intensive Care, Department of Medicine - DIMED, Padua University Hospital, Via Cesare Battisti 267, 35128, Padua, Italy
| | - Karin Boer
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Myrthe van Baardwijk
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Flavia Neri
- Kidney and Pancreas Transplantation Unit, Department of Surgical, Oncological and Gastroenterological Sciences, Padua University Hospital, Via Giustiniani 2, 35128, Padua, Italy
| | - Manuela Campisi
- Occupational Medicine, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padua University, Via Giustiniani 2, 35128, Padua, Italy
| | - Hendrikus J A N Kimenai
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Marian C Clahsen-van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Sofia Pavanello
- Occupational Medicine, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padua University, Via Giustiniani 2, 35128, Padua, Italy
| | - Lucrezia Furian
- Kidney and Pancreas Transplantation Unit, Department of Surgical, Oncological and Gastroenterological Sciences, Padua University Hospital, Via Giustiniani 2, 35128, Padua, Italy
| | - Robert C Minnee
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| |
Collapse
|
3
|
Blinova VG, Gladilina YA, Eliseeva DD, Lobaeva TA, Zhdanov DD. [Increased suppressor activity of transformed ex vivo regulatory T-cells in comparison with unstimulated cells of the same donor]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:55-67. [PMID: 35221297 DOI: 10.18097/pbmc20226801055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Regulatory T-cells CD4⁺CD25⁺FoxP3⁺CD127low (Tregs) play a key role in the maintenance of tolerance to auto antigens, inhibit function of effector T and B lymphocytes, and provide a balance between effector and regulatory arms of immunity. Patients with autoimmune diseases have decreased Treg numbers and impaired suppressive activity. Transformed ex vivo autologous Tregs could restore destroyed balance of the immune system. We developed a method for Treg precursor cell cultivation. Following the method, we were able to grown up 300-400 million of Tregs cells from 50 ml of peripheral blood during a week. Transformed ex vivo Tregs are 90-95% CD4⁺CD25⁺FoxP3⁺CD127low and have increased expression of transcription genes FoxP3 and Helios. Transformed ex vivo Tregs have increased demethylation of FoxP3 promoter and activated genes of proliferation markers Cycline B1, Ki67 and LGALS 1. Transformed ex vivo Tregs have increased suppressive activity and up to 80-90% these cells secrete cytokines TNFα и IFNγ. Our data suggest transformed ex vivo autologous Tregs have genetic, immunophenotypic and functional characteristics for regulatory T-cells and further can be used for adoptive immunotherapy autoimmune diseases and inhibition of transplantation immunity.
Collapse
Affiliation(s)
- V G Blinova
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | - T A Lobaeva
- Department of Biochemistry, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
| | - D D Zhdanov
- Institute of Biomedical Chemistry, Moscow, Russia; Department of Biochemistry, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
4
|
AKGÖLLÜ E. Effect of FOXP3 gene variants on the immune-active HBV and inactive HBV phases. CUKUROVA MEDICAL JOURNAL 2021. [DOI: 10.17826/cumj.978353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
5
|
Lai X, Yao Z, Ning F, Zhang L, Fang J, Li G, Xu L, Xiong Y, Liu L, Chen R, Ma J, Chen Z. Blockade of OX40/OX40L pathway combined with ethylene-carbodiimide-fixed donor splenocytes induces donor-specific allograft tolerance in presensitized recipients. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:84. [PMID: 32175377 DOI: 10.21037/atm.2019.12.146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Memory T cells (Tms) are the major barrier preventing long-term allograft survival in presensitized transplant recipients. The OX40/OX40L pathway is important in the induction and maintenance of Tms. Methods In this study, we added anti-OX40L mAb to ethylene-carbodiimide-fixed donor splenocytes (ECDI-SPs)-a method which is effective in inducing allograft tolerance in non-presensitized mouse heart transplant model. Recipient mice received heart transplantation after 6 weeks of donor skin presensitization and were treated with anti-OX40L mAb, ECDI-SPs or anti-OX40L mAb + ECDI-SPs, respectively. Results Our data showed that the combination of ECDI-SPs and anti-OX40L mAb induced donor-specific tolerance in skin-presensitized heart transplant recipients, with the mechanism for this being associated with suppression of Tms and upregulation of CD4+CD25+Foxp3+ T regulatory cells (Tregs). Importantly, CD25+ T-cell depletion in the combined therapy-treated recipients broke the establishment of allograft tolerance, whereas adoptive transfer of presensitization-derived T cells into tolerant recipients suppressed Tregs expansion and abolished established tolerance. Conclusions Blockade of OX40/OX40L pathway in combination with ECDI-SPs appears to modulate the Tms/Tregs imbalance so as to create a protective milieu and induce graft tolerance in presensitized recipients.
Collapse
Affiliation(s)
- Xingqiang Lai
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zhongpeng Yao
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Fen Ning
- Guangzhou Institute of Pediatrics, Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Lei Zhang
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jiali Fang
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Guanghui Li
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Lu Xu
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yunyi Xiong
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Luhao Liu
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Rongxin Chen
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Junjie Ma
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zheng Chen
- Organ Transplant Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
6
|
Akgöllü E. Evaluation of Forkhead Box P3 gene polymorphisms in chronic HBV infection. J Gene Med 2020; 22:e3172. [PMID: 32037623 DOI: 10.1002/jgm.3172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/15/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) infection causes liver failure, liver cirrhosis and hepatocellular carcinoma. The FOXP3 gene polymorphisms, the rs2232365 A/G and the rs3761548 A/C, were identified to be associated with regulatory T cell-mediated immunosuppression. The response to HBV infection may be affected by FOXP3 polymorphisms. The present study aimed to assess the relationship between FOXP3 gene polymorphisms and chronic HBV infection risk. METHODS FOXP3 gene polymorphisms were explored in 237 chronic HBV patients and in 237 individuals with HBV spontaneous clearance using a real-time polymerase chain reaction. RESULTS The patients with rs2232365 AG and rs3761548 AC genotype had a 1.20- and a 1.58-fold greater HBV risk than non-carriers patients, although they were not significant. Moreover, the AA genotypes of both polymorphisms in the males and females had an increased the persistent HBV risk, although this also was not statistically significant. CONCLUSIONS In conclusion, the present study is the first report to demonstrate that these polymorphisms have no effect on the risk of chronic HBV infection. This results suggest that FOXP3 gene polymorphisms and FOXP3 expression should be evaluated together with frequency of regulatory T cells in HBV infection.
Collapse
Affiliation(s)
- Ersin Akgöllü
- Department of Gastroenterology, Faculty of Medicine, Çukurova University, Adana, Turkey
| |
Collapse
|
7
|
Contact-independent suppressive activity of regulatory T cells is associated with telomerase inhibition, telomere shortening and target lymphocyte apoptosis. Mol Immunol 2018; 101:229-244. [PMID: 30025223 DOI: 10.1016/j.molimm.2018.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/02/2018] [Accepted: 07/11/2018] [Indexed: 02/08/2023]
Abstract
Regulatory T cells (Tregs) play a fundamental role in the maintenance of immunological tolerance by suppressing effector target T, B and NK lymphocytes. Contact-dependent suppression mechanisms have been well-studied, though contact-independent Treg activity is not fully understood. In the present study, we showed that human native Tregs, as well as induced ex vivo Tregs, can cause in vitro telomere-dependent senescence in target T, B and NK cells in a contact-independent manner. The co-cultivation of target cells with Tregs separated through porous membranes induced alternative splicing of the telomerase catalytic subunit hTERT (human Telomerase Reverse Transcriptase), which suppressed telomerase activity. Induction of the hTERT splicing variant was associated with increased expression of the apoptotic endonuclease EndoG, a splicing regulator. Inhibited telomerase in target cells co-cultivated with Tregs for a long period of time led to a decrease in their telomere lengths, cell cycle arrest, conversion of the target cells to replicative senescence and apoptotic death. Induced Tregs showed the ability to up-regulate EndoG expression, TERT alternative splicing and telomerase inhibition in mouse T, B and NK cells after in vivo administration. The results of the present study describe a novel mechanism of contact-independent Treg cell suppression that induces telomerase inhibition through the EndoG-provoked alternative splicing of hTERT and converts cells to senescence and apoptosis phenotypes.
Collapse
|
8
|
A Verghese D, Demir M, Chun N, Fribourg M, Cravedi P, Llaudo I, Woodruff TM, Yadav P, Lira SA, Medof ME, Heeger PS. T Cell Expression of C5a Receptor 2 Augments Murine Regulatory T Cell (T REG) Generation and T REG-Dependent Cardiac Allograft Survival. THE JOURNAL OF IMMUNOLOGY 2018; 200:2186-2198. [PMID: 29436411 DOI: 10.4049/jimmunol.1701638] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/11/2018] [Indexed: 12/26/2022]
Abstract
C5aR2 (C5L2/gp77) is a seven-transmembrane spanning receptor that binds to C5a but lacks motifs essential for G protein coupling and associated signal transduction. C5aR2 is expressed on immune cells, modulates various inflammatory diseases in mice, and has been shown to facilitate murine and human regulatory T cell (TREG) generation in vitro. Whether and how C5aR2 impacts in vivo TREG generation and pathogenic T cell-dependent disease models have not been established. In this article, we show that murine T cells express and upregulate C5aR2 during induced TREG (iTREG) generation and that the absence of T cell-expressed C5aR2 limits in vivo iTREG generation following adoptive transfer of naive CD4+ T cells into Rag1-/- recipients. Using newly generated C5aR2-transgenic mice, we show that overexpression of C5aR2 in naive CD4+ T cells augments in vivo iTREG generation. In a model of TREG-dependent cardiac allograft survival, recipient C5aR2 deficiency accelerates graft rejection associated with lower TREG/effector T cell ratios, whereas overexpression of C5aR2 in immune cells prolongs graft survival associated with an increase in TREG/effector T cell ratios. T cell-expressed C5aR2 modulates TREG induction without altering effector T cell proliferation or cytokine production. Distinct from reported findings in neutrophils and macrophages, TREG-expressed C5aR2 does not interact with β-arrestin or inhibit ERK1/2 signaling. Rather, cumulative evidence supports the conclusion that C5aR2 limits C5aR1-initiated signals known to inhibit TREG induction. Together, the data expand the role of C5aR2 in adaptive immunity by providing in vivo evidence that T cell-expressed C5aR2 physiologically modulates iTREG generation and iTREG-dependent allograft survival.
Collapse
Affiliation(s)
- Divya A Verghese
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Markus Demir
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Nicholas Chun
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Miguel Fribourg
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Paolo Cravedi
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ines Llaudo
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane St. Lucia, Brisbane, Queensland 4072, Australia; and
| | - Pragya Yadav
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sergio A Lira
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - M Edward Medof
- Institute of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Peter S Heeger
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029; .,Translational Transplant Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
9
|
Vilchez V, Turcios L, Butterfield DA, Mitov MI, Coquillard CL, Brandon JA, Cornea V, Gedaly R, Marti F. Evidence of the immunomodulatory role of dual PI3K/mTOR inhibitors in transplantation: an experimental study in mice. Transpl Int 2017; 30:1061-1074. [DOI: 10.1111/tri.12989] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Valery Vilchez
- Department of Surgery; College of Medicine; University of Kentucky; Lexington KY USA
| | - Lilia Turcios
- Department of Surgery; College of Medicine; University of Kentucky; Lexington KY USA
| | - David A. Butterfield
- Redox Metabolism (RM) Shared Resource Facility (SRF); Markey Cancer Center; College of Medicine; University of Kentucky; Lexington KY USA
- Department of Chemistry; College of Medicine; University of Kentucky; Lexington KY USA
| | - Mihail I. Mitov
- Redox Metabolism (RM) Shared Resource Facility (SRF); Markey Cancer Center; College of Medicine; University of Kentucky; Lexington KY USA
| | - Cristin L. Coquillard
- Department of Surgery; College of Medicine; University of Kentucky; Lexington KY USA
| | - Ja Anthony Brandon
- Department of Internal Medicine; College of Medicine; University of Kentucky; Lexington KY USA
| | - Virgilius Cornea
- Department of Pathology and Laboratory Medicine; College of Medicine; University of Kentucky; Lexington KY USA
| | - Roberto Gedaly
- Department of Surgery; College of Medicine; University of Kentucky; Lexington KY USA
| | - Francesc Marti
- Department of Surgery; College of Medicine; University of Kentucky; Lexington KY USA
| |
Collapse
|
10
|
Splenocyte Infusion and Whole-Body Irradiation for Induction of Peripheral Tolerance in Porcine Lung Transplantation: Modifications of the Preconditioning Regime for Improved Clinical Feasibility. Transplant Direct 2017; 3:e170. [PMID: 28706973 PMCID: PMC5498011 DOI: 10.1097/txd.0000000000000689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/20/2017] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Preoperative low-dose whole-body irradiation (IRR) with 1.5 and 7 Gy thymic IRR of the recipient, combined with a perioperative donor splenocyte infusion lead to reliable donor specific peripheral tolerance in our allogeneic porcine lung transplantation model. To reduce the toxicity of this preconditioning regime, modifications of the IRR protocol and their impact on allograft survival were assessed. METHODS Left-sided single lung transplantation from major histocompatibility complex and sex mismatched donors was performed in 14 adult female minipigs. Recipient animals were exposed to 3 different protocols of nonmyeloablative IRR within 12 hours before transplantation. All animals were administered a donor splenocyte infusion on the day of lung transplantation. Intravenous pharmacologic immunosuppression was withdrawn after 28 postoperative days. Allograft survival was monitored by chest radiographs and bronchoscopy. RESULTS IRR prolonged transplant survival in a dose- and field-dependent manner. Shielding of the bone marrow from IRR (total lymphoid IRR at 1.5 and 7 Gy thymic IRR) significantly reduced protocol toxicity defined as thrombocytopenia and consecutive increased bleeding propensity, but had a less effective impact on graft survival. Whole-body IRR at 0.5 and 7 Gy thymic IRR proved to be ineffective for reliable tolerance induction. Eventually, high levels of circulating CD4+CD25high regulatory T cells were present in long-term survivors. CONCLUSIONS These data show that the infusion of donor-specific alloantigen in combination with IRR is efficient once a threshold dose is exceeded.
Collapse
|
11
|
Shatrova AN, Mityushova EV, Vassilieva IO, Aksenov ND, Zenin VV, Nikolsky NN, Marakhova II. Time-Dependent Regulation of IL-2R α-Chain (CD25) Expression by TCR Signal Strength and IL-2-Induced STAT5 Signaling in Activated Human Blood T Lymphocytes. PLoS One 2016; 11:e0167215. [PMID: 27936140 PMCID: PMC5172478 DOI: 10.1371/journal.pone.0167215] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/10/2016] [Indexed: 12/30/2022] Open
Abstract
The expression of the IL-2R α-chain (IL-2Rα) is regulated at the transcriptional
level via TCR- and IL-2R-signaling. The question is how to precede in time the
activation signals to induce the IL-2Rα expression in native primary T cells. By
comparing the effects of selective drugs on the dynamics of CD25 expression
during the mitogen stimulation of human peripheral blood lymphocytes, we
identified distinct Src- and JAK-dependent stages of IL-2Rα upregulation. PP2, a
selective inhibitor of TCR-associated Src kinase, prevents CD25 expression at
initial stages of T cell activation, prior to the cell growth. This early IL-2Rα
upregulation underlies the T cell competence and the IL-2 responsiveness. We
found that the activated with “weak” mitogen, the population of blood
lymphocytes has some pool of competent CD25+ cells bearing a high affinity
IL-2R. A distinct pattern of IL-2R signaling in resting and competent T
lymphocytes has been shown. Based on the inhibitory effect of WHI-P131, a
selective drug of JAK3 kinase activity, we concluded that in quiescent primary T
lymphocytes, the constitutive STAT3 and the IL-2-induced prolonged STAT5
activity (assayed by tyrosine phosphorylation) is mostly JAK3-independent. In
competent T cells, in the presence of IL-2 JAK3/STAT5 pathway is switched to
maintain the higher and sustained IL-2Rα expression as well as cell growth and
proliferation. We believe that understanding the temporal coordination of
antigen- and cytokine-evoked signals in primary T cells may be useful for
improving immunotherapeutic strategies.
Collapse
Affiliation(s)
- Alla N. Shatrova
- Department of Intracellular Signaling and Transport,
Institute of Cytology, Russian Academy of Sciences, St-Petersburg,
Russia
| | - Elena V. Mityushova
- Department of Intracellular Signaling and Transport,
Institute of Cytology, Russian Academy of Sciences, St-Petersburg,
Russia
| | - Irina O. Vassilieva
- Department of Intracellular Signaling and Transport,
Institute of Cytology, Russian Academy of Sciences, St-Petersburg,
Russia
| | - Nikolay D. Aksenov
- Department of Intracellular Signaling and Transport,
Institute of Cytology, Russian Academy of Sciences, St-Petersburg,
Russia
| | - Valery V. Zenin
- Department of Intracellular Signaling and Transport,
Institute of Cytology, Russian Academy of Sciences, St-Petersburg,
Russia
| | - Nikolay N. Nikolsky
- Department of Intracellular Signaling and Transport,
Institute of Cytology, Russian Academy of Sciences, St-Petersburg,
Russia
| | - Irina I. Marakhova
- Department of Intracellular Signaling and Transport,
Institute of Cytology, Russian Academy of Sciences, St-Petersburg,
Russia
- * E-mail:
| |
Collapse
|
12
|
Glycogen synthase kinase 3β inhibition promotes human iTreg differentiation and suppressive function. Immunol Res 2016; 62:60-70. [PMID: 25752456 DOI: 10.1007/s12026-015-8635-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced regulatory T cells (iTregs) are essential to maintain immunological tolerance, immune homeostasis and prevention of autoimmunity. Some studies suggest that glycogen synthase kinase 3β (GSK3β) is involved in the mouse iTreg differentiation; however, whether GSK3β inhibits or enhances iTreg differentiation is still a matter of controversy. To address this issue, we have utilized human naïve CD4(+) T cells and investigated whether GSK3 activity changes during iTreg differentiation and whether altering GSK3 activity influences the development of iTregs and its suppressive function. As a constitutively activated kinase, during iTreg differentiation GSK3β became quickly deactivated (phosphorylated at serine 9), which is dependent on MAPK pathway rather than PI3-kinase/Akt pathway. Our results indicated that inhibition of GSK3β by specific inhibitors, SB216763 or TDZD-8, promoted the differentiation of iTreg and increased their suppressive activity. In contrast, overexpression of GSK3β significantly inhibited iTreg differentiation. Furthermore, GSK3β inhibition enhanced iTreg differentiation through the TGF-β/Smad3 pathway. Taken together, this study demonstrates that inhibition of GSK3β enhances human iTreg differentiation and its suppressive activity, and provides a rationale to target GSK3β as a novel immunotherapeutic strategy.
Collapse
|
13
|
Effect of FOXP3 polymorphism on the clinical outcomes after allogeneic hematopoietic stem cell transplantation in pediatric acute leukemia patients. Int Immunopharmacol 2015; 31:132-9. [PMID: 26735609 DOI: 10.1016/j.intimp.2015.12.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/15/2015] [Accepted: 12/17/2015] [Indexed: 12/15/2022]
Abstract
Forkhead BOX P3 (FOXP3) polymorphisms have recently been investigated as candidate risk factors in several tumors and autoimmune diseases. This study aims to evaluate the potential influence of FOXP3 rs3761548 polymorphism in the donor on the outcome of allogeneic hematopoietic stem cell transplantation (allo-HSCT). A total of 171 patients were enrolled for this study and genotyped using direct sequencing. Patients with rs3761548 CC genotype had higher incidence of hepatic veno-occlusive disease (HVOD) and cytomegalovirus (CMV) infection than that of the individuals with AA or AC genotype (P=0.011, P=0.023). Treatment-related mortality (TRM) rate of patients with AA or AC genotype was lower than that of the patients with CC genotype (P=0.044) resulting in a difference in overall survival (OS). However, there was no difference in graft-versus-host disease (GVHD) relapse or blood stream infection (BSI), depending on the genotype at rs3761548 locus. In multivariate analysis, CC genotype showed as a risk factor in the development of HVOD and CMV infection, with low OS. In conclusion, this is the first report on FOXP3 rs3761548 SNP in allo-HSCT and we suggest that this SNP be considered a candidate marker for predicting the development of HVOD and CMV infection after allo-HSCT.
Collapse
|
14
|
Fan CB, Wang Y, Wang QP, Du KL, Wen DG, Ouyang J. Alloantigen-specific T-cell hyporesponsiveness induced by dnIKK2 gene-transfected recipient immature dendritic cells. Cell Immunol 2015; 297:100-7. [DOI: 10.1016/j.cellimm.2015.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 07/26/2015] [Accepted: 07/31/2015] [Indexed: 01/13/2023]
|
15
|
Dai H, Peng F, Lin M, Xia J, Yu S, Lan G, Wang Y, Xie X, Fang C, Corbascio M, Qi Z, Peng L. Anti-OX40L monoclonal antibody prolongs secondary heart allograft survival based on CD40/CD40L and LFA-1/ICAM-1 blockade. Transpl Immunol 2015; 32:84-91. [PMID: 25613092 DOI: 10.1016/j.trim.2015.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/11/2015] [Accepted: 01/12/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Memory T cells (Tms) form a barrier against long-term allograft survival; however, CD4(+)Foxp3(+) regulatory T cells (Tregs) can suppress allograft rejection. The OX40/OX40L pathway is critical to the generation of Tms and turns off Treg suppressor function. METHODS B6 mice that rejected BALB/c skin grafts after 4 weeks were used as the secondary heart transplant recipients. The skin recipient mice, termed S0, S2 and S3, were treated with the isotype antibodies, anti-CD40L/LFA-1 or anti-OX40L combined with anti-CD40L/LFA-1 mAbs, respectively. The secondary heart recipients, termed H0 and H2, received anti-CD40L/LFA-1 mAbs or not, respectively (Fig. 1). RESULTS Four weeks after primary skin transplantation, the Tms in the S3 group that received anti-OX40L with anti-CD40L/LFA-1 mAbs were reduced compared to those in the S2 group (CD4(+) Tm: 32.61 ± 2.20% in S2 vs. 25.36 ± 1.16% in S3; CD8(+) Tm: 27.76 ± 1.96% in S2 vs. 20.95 ± 1.30% in S3; P < 0.01). Meanwhile, the proportions of Tregs in S3 increased compared to those in S2 (P < 0.05). The anti-OX40L with anti-CD40L/LFA-1 mAbs group (S3H2) prolonged the mean survival time (MST) following secondary heart transplantation from 9.5 days to 21 days (P < 0.001). Furthermore, allogeneic proliferation of recipient splenic T cells and graft-infiltrating lymphocytes were significantly inhibited in the S3H2 group. Additionally, a higher level of IL-10 was detected in sera and allografts. CONCLUSIONS Anti-OX40L mAb could prolong secondary heart allograft survival based on CD40/CD40L and LFA-1/ICAM-1 blockade. The mechanism of protecting allografts using anti-OX40L mAb involved impairing the generation of Tm and up-regulating IL-10 producing Tregs, inhibiting the function of T cells.
Collapse
Affiliation(s)
- Helong Dai
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Fenghua Peng
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Minjie Lin
- Department of Cardiology, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Junjie Xia
- Organ Transplantation Institute, Xiamen University, Fujian Province, PR China
| | - Shaojie Yu
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Gongbin Lan
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Yu Wang
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Xubiao Xie
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Chunhua Fang
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | | | - Zhongquan Qi
- Organ Transplantation Institute, Xiamen University, Fujian Province, PR China
| | - Longkai Peng
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China.
| |
Collapse
|
16
|
Ohta A, Sitkovsky M. Extracellular adenosine-mediated modulation of regulatory T cells. Front Immunol 2014; 5:304. [PMID: 25071765 PMCID: PMC4091046 DOI: 10.3389/fimmu.2014.00304] [Citation(s) in RCA: 234] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 06/15/2014] [Indexed: 12/20/2022] Open
Abstract
Extracellular adenosine-dependent suppression and redirection of pro-inflammatory activities are mediated by the signaling through adenosine receptors on the surface of most immune cells. The immunosuppression by endogenously-produced adenosine is pathophysiologically significant since inactivation of A2A/A2B adenosine receptor (A2AR/A2BR) and adenosine-producing ecto-enzymes CD39/CD73 results in the higher intensity of immune response and exaggeration of inflammatory damage. Regulatory T cells (Treg) can generate extracellular adenosine, which is implicated in the immunoregulatory activity of Tregs. Interestingly, adenosine has been shown to increase the numbers of Tregs and further promotes their immunoregulatory activity. A2AR-deficiency in Tregs reduces their immunosuppressive efficacy in vivo. Thus, adenosine is not only directly and instantly inhibiting to the immune response through interaction with A2AR/A2BR on the effector cells, but also adenosine signaling can recruit other immunoregulatory mechanisms, including Tregs. Such interaction between adenosine and Tregs suggests the presence of a positive feedback mechanism, which further promotes negative regulation of immune system through the establishment of immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Akio Ohta
- New England Inflammation and Tissue Protection Institute, Northeastern University , Boston, MA , USA
| | - Michail Sitkovsky
- New England Inflammation and Tissue Protection Institute, Northeastern University , Boston, MA , USA
| |
Collapse
|
17
|
Mikami N, Sakaguchi S. CD28 signals the differential control of regulatory T cells and effector T cells. Eur J Immunol 2014; 44:955-7. [PMID: 24652756 DOI: 10.1002/eji.201444513] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 02/27/2014] [Accepted: 03/17/2014] [Indexed: 01/08/2023]
Abstract
One possible means of driving antigen-specific immune suppression is to expand or induce antigen-specific FoxP3-expressing Treg cells. One way of activating and expanding these specialized cells, both in vitro and in vivo, is by strong costimulation via CD28 with an agonistic anti-CD28 monoclonal antibody, called anti-CD28 superagonist (CD28SA). However, CD28SA also strongly activates conventional T (Tconv) cells to secrete proinflammatory cytokines and, under certain conditions, causes serious cytokine release syndrome. In this issue of European Journal of Immunology, Tabares et al. [Eur. J. Immunol. 2014. 44: 1225-1236] address how CD28SA can be used for the differential control of human Treg and Tconv cells to suppress immune responses without serious adverse effects. They show that, depending on the dose of the antibody or by comedication of cortico-steroid, the selective expansion of Treg cells can be achieved without significantly activating Tconv cells to produce inflammatory cytokines. This difference in CD28 signal sensitivity between the two populations can be exploited for better control of immune responses.
Collapse
Affiliation(s)
- Norihisa Mikami
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | | |
Collapse
|
18
|
Dugast E, Chesneau M, Soulillou JP, Brouard S. Biomarkers and possible mechanisms of operational tolerance in kidney transplant patients. Immunol Rev 2014; 258:208-17. [DOI: 10.1111/imr.12156] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Emilie Dugast
- INSERM UMR 1064; Nantes France
- Centaure; Nantes France
| | - Mélanie Chesneau
- INSERM UMR 1064; Nantes France
- Université de Nantes; Nantes France
| | - Jean-Paul Soulillou
- INSERM UMR 1064; Nantes France
- Centaure; Nantes France
- CHU de Nantes; Nantes France
- Université de Nantes; Nantes France
| | - Sophie Brouard
- INSERM UMR 1064; Nantes France
- Centaure; Nantes France
- CHU de Nantes; Nantes France
- Université de Nantes; Nantes France
| |
Collapse
|