51
|
Kollar B, Kamat P, Klein H, Waldner M, Schweizer R, Plock J. The Significance of Vascular Alterations in Acute and Chronic Rejection for Vascularized Composite Allotransplantation. J Vasc Res 2019; 56:163-180. [DOI: 10.1159/000500958] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/14/2019] [Indexed: 11/19/2022] Open
|
52
|
Treg-mediated prolonged survival of skin allografts without immunosuppression. Proc Natl Acad Sci U S A 2019; 116:13508-13516. [PMID: 31196957 PMCID: PMC6613183 DOI: 10.1073/pnas.1903165116] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Injection of Interleukin-2 (IL-2) complexed with a particular anti-IL-2 monoclonal antibody (mab) JES6-1 has been shown to selectively expand CD4+Foxp3+ T regulatory T cells (Tregs) in vivo. Although the potency of this approach with regard to transplantation has already been proven in an islet transplantation model, skin graft survival could not be prolonged. Since the latter is relevant to human allograft survival, we sought to improve the efficiency of IL-2 complex (cplx) treatment for skin allograft survival in a stringent murine skin graft model. Here, we show that combining low doses of IL-2 cplxs with rapamycin and blockade of the inflammatory cytokine IL-6 leads to long-term (>75 d) survival of major histocompatibility complex-different skin allografts without the need for immunosuppression. Allograft survival was critically dependent on CD25+FoxP3+ Tregs and was not accompanied by impaired responsiveness toward donor alloantigens in vitro after IL-2 cplx treatment was stopped. Furthermore, second donor-type skin grafts were rejected and provoked rejection of the primary graft, suggesting that operational tolerance is not systemic but restricted to the graft. These findings plus the lack of donor-specific antibody formation imply that prolonged graft survival was largely a reflection of immunological ignorance. The results may represent a potentially clinically translatable strategy for the development of protocols for tolerance induction.
Collapse
|
53
|
Boroughs AC, Larson RC, Choi BD, Bouffard AA, Riley LS, Schiferle E, Kulkarni AS, Cetrulo CL, Ting D, Blazar BR, Demehri S, Maus MV. Chimeric antigen receptor costimulation domains modulate human regulatory T cell function. JCI Insight 2019; 5:126194. [PMID: 30869654 PMCID: PMC6538349 DOI: 10.1172/jci.insight.126194] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 03/11/2019] [Indexed: 02/06/2023] Open
Abstract
Regulatory T cells (Tregs) are key modulators of inflammation and are important for the maintenance of peripheral tolerance. Adoptive immunotherapy with polyclonal Tregs holds promise in organ transplantation, graft-versus-host disease, and autoimmune diseases, but may be enhanced by antigen-specific, long-lived Treg cells. We modified primary human Tregs with chimeric antigen-receptors (CARs) bearing different costimulatory domains and performed in vitro analyses of their phenotype and function. While neither the presence of a CAR nor the type of costimulation domain influenced Foxp3 expression in Tregs, the costimulation domain of the CARs affected CAR Treg surface phenotype and functions such as cytokine production. Furthermore, signaling from the CD28 costimulation domain maintained CAR Treg suppressor function, whereas 4-1B costimulation did not. In vivo, CAR Tregs accumulated at sites expressing target antigen, and suppressed antigen specific effector T cell responses; however, only CAR Tregs with CD28 signaling domains were potent inhibitors of effector T cell mediated graft rejection in vivo. Our findings support the use of CD28 based CAR-Tregs for tissue specific immune suppression in the clinic.
Collapse
Affiliation(s)
- Angela C. Boroughs
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Immunology Program, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca C. Larson
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Immunology Program, Harvard Medical School, Boston, Massachusetts, USA
| | - Bryan D. Choi
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurosurgery and
| | - Amanda A. Bouffard
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lauren S. Riley
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Erik Schiferle
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | - David Ting
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Bruce R. Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shadmehr Demehri
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Marcela V. Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Massachusetts, USA
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
54
|
Transplant arteriosclerosis in humanized mice reflects chronic lung allograft dysfunction and is controlled by regulatory T cells. J Thorac Cardiovasc Surg 2019; 157:2528-2537. [PMID: 30955963 DOI: 10.1016/j.jtcvs.2019.01.134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 01/04/2019] [Accepted: 01/06/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Chronic lung allograft dysfunction (CLAD) is a severe complication of lung transplantation limiting long-term survival. We studied correlations between CLAD after clinical lung transplantation and leukocyte-mediated development of transplant arteriosclerosis (TA) in a humanized mouse model. The pericardiophrenic artery was procured from surplus tissue of donor lungs (n = 22) transplanted in our clinical program and was implanted into the abdominal aorta of immune-deficient mice. METHODS Allogeneic human peripheral blood mononuclear cells (PBMCs) had been procured 1 day after lung transplantation from the respective recipients with or without enriching for CD4+CD25high T cells were used. TA was assessed in mice 28 days later by histology. The respective clinical lung recipients were later divided into 2 groups. Eight patients (36.3%) had developed CLAD 23 ± 5 months after lung transplantation, whereas the remaining 14 (63.6%) did not develop CLAD within 25 ± 5 months. RESULTS In the PBMC CLAD+ group of mouse experiments, TA was significantly more severe than in the PBMC CLAD- group (39.9% ± 13% vs 14.9% ± 4% intimal thickening; P = .0081). Then, intimal thickening was significantly inhibited in the PBMC+ regulatory T cells CLAD+ group compared with the PBMC CLAD+ group (0.4% ± 4% vs 39.9% ± 13%; P = .003). In the experiments using PBMCs from lung recipients without CLAD, enriching regulatory T cells also suppressed the development of TA (0.9% ± 3% PBMC CLAD- vs 14.9% ± 4% PBMC+ regulatory T cells CLAD-; P = .001). CONCLUSIONS Lung transplant recipients who later develop CLAD have peripheral leukocytes already at the time of transplant that transfer proinflammatory properties leading to TA in a humanized mouse model. TA remains sensitive to inhibition by autologous regulatory T cells, suggesting a cell therapy-based approach for the prevention of CLAD after lung transplantation.
Collapse
|
55
|
Issa F, Milward K, Goto R, Betts G, Wood KJ, Hester J. Transiently Activated Human Regulatory T Cells Upregulate BCL-XL Expression and Acquire a Functional Advantage in vivo. Front Immunol 2019; 10:889. [PMID: 31068951 PMCID: PMC6491764 DOI: 10.3389/fimmu.2019.00889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/05/2019] [Indexed: 01/08/2023] Open
Abstract
Regulatory T cells (Tregs) can control excessive or undesirable immune responses toward autoantigens, alloantigens, and pathogens. In transplantation, host immune responses against the allograft are suppressed through the use of immunosuppressive drugs, however this often results in life-threatening side effects including nephrotoxicity and an increased incidence of cancer and opportunistic infections. Tregs can control graft-vs.-host disease and transplant rejection in experimental models, providing impetus for the use of Tregs as a cellular therapy in clinical transplantation. One of the major barriers to the widespread use of Treg cellular therapy is the requirement to expand cells ex vivo to large numbers in order to alter the overall balance between regulatory and effector cells. Methods that enhance suppressive capacity thereby reducing the need for expansion are therefore of interest. Here, we have compared the function of freshly-isolated and ex vivo-manipulated human Tregs in a pre-clinical humanized mouse model of skin transplantation. Sorted human CD127loCD25+CD4+ Tregs were assessed in three different conditions: freshly-isolated, following transient in vitro activation with antiCD3/antiCD28 beads or after ex vivo-expansion for 2 weeks in the presence of antiCD3/antiCD28 beads and recombinant human IL2. While ex vivo-expansion of human Tregs increased their suppressive function moderately, transient in vitro-activation of freshly isolated Tregs resulted in a powerful enhancement of Treg activity sufficient to promote long-term graft survival of all transplants in vivo. In order to investigate the mechanisms responsible for these effects, we measured the expression of Treg-associated markers and susceptibility to apoptosis in activated Tregs. Transiently activated Tregs displayed enhanced survival and proliferation in vitro and in vivo. On a molecular level, Treg activation resulted in an increased expression of anti-apoptotic BCL2L1 (encoding BCL-XL) which may be at least partially responsible for the observed enhancement in function. Our results suggest that in vitro activation of human Tregs arms them with superior proliferative and survival abilities, enabling them to more effectively control alloresponses. Importantly, this transient activation results in a rapid functional enhancement of freshly-isolated Tregs, thereby providing an opportunity to eliminate the need for in vitro expansion in select circumstances. A protocol employing this technique would therefore benefit from a reduced requirement for large cell numbers for effective therapy.
Collapse
|
56
|
Abstract
In this chapter, we describe the history of transplantation, the multiple cell types, and mechanisms that are involved in rejection and tolerance of a transplanted organ, as well as summarize the common and promising new therapeutics used in transplant patients.
Collapse
Affiliation(s)
- Jessica Stolp
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Masaaki Zaitsu
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
57
|
Wedel J, Bruneau S, Liu K, Kong SW, Sage PT, Sabatini DM, Laplante M, Briscoe DM. DEPTOR modulates activation responses in CD4 + T cells and enhances immunoregulation following transplantation. Am J Transplant 2019; 19:77-88. [PMID: 29969188 PMCID: PMC6310634 DOI: 10.1111/ajt.14995] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 01/25/2023]
Abstract
DEPTOR is an evolutionarily conserved cell-intrinsic binding partner of mTOR that functions as a negative regulator of signaling responses. In this study, we show that DEPTOR is expressed within CD4+ T cells, and we observed that its relative level of expression modulates differentiation as well as glucose utilization within CD4+ T effectors in vitro. Using knock-in mice, we also find that induced expression of DEPTOR within CD4+ T regulatory cells stabilizes Foxp3 expression, shifts metabolism toward oxidative phosphorylation, and increases survival and suppressive function. In vivo, fully MHC mismatched cardiac allograft survival is significantly prolonged in knock-in recipients and sustained recipient expression of DEPTOR in combination with costimulatory blockade induces long-term graft survival. Furthermore, we show that the induced expression of DEPTOR in CD4+ T effectors fails to inhibit acute allograft rejection. Rather, prolonged survival is dominantly mediated via induced expression and function of DEPTOR within recipient CD4+ T regulatory cells. These collective findings identify DEPTOR as a novel protein that functions in CD4+ T cells to augment immunoregulation in vitro and in vivo.
Collapse
Affiliation(s)
- Johannes Wedel
- Transplant Research Program, Boston Children’s Hospital, Boston, MA 02115, USA.,Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Boston, MA 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah Bruneau
- Transplant Research Program, Boston Children’s Hospital, Boston, MA 02115, USA.,Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Boston, MA 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Kaifeng Liu
- Transplant Research Program, Boston Children’s Hospital, Boston, MA 02115, USA.,Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Boston, MA 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Sek Won Kong
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.,Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Peter T. Sage
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA 02115, USA
| | - David M. Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Cambridge, MA 02142, USA
| | - Mathieu Laplante
- Centre de researche de l’Institut Universitaire de Cardiologie et de Pulmonologie de Quebec, Universite Lavel, Faculte de Medecine, Quebec, QC, Canada
| | - David M. Briscoe
- Transplant Research Program, Boston Children’s Hospital, Boston, MA 02115, USA.,Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Boston, MA 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.,Corresponding author: David M. Briscoe, M.D.,
| |
Collapse
|
58
|
Milward K, Hester J, Wood KJ. Isolation of Human Regulatory T Lymphocytes by Fluorescence-Activated Cell Sorting. Methods Mol Biol 2019; 1899:43-54. [PMID: 30649764 DOI: 10.1007/978-1-4939-8938-6_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Regulatory T cells (Tregs) are a population of lymphocytes that exerts suppressive effects upon the immune system. In human peripheral blood, the major population of T lymphocytes with suppressive capacity are defined by expression of the T cell co-receptor CD4 and the interleukin-2 receptor α-chain (CD25), combined with minimal expression of the interleukin-7 receptor α subunit (CD127). We begin by outlining the method for isolating peripheral blood mononuclear cells (PBMCs) from human blood by centrifugation of whole blood overlayed on a hydrophilic polysaccharide, with an additional erythrocyte lysis step. The protocol that follows utilizes Fluorescence-Activated Cell Sorting (FACS) for the isolation of this CD4+CD25+CD127lo population of regulatory T cells, with high yield and purity, from immunostained PBMCs. Prior to FACS isolation, this protocol exploits magnetic immunoselection for pre-enrichment of CD25+ PBMC, which reduces the duration of the subsequent FACS isolation.
Collapse
Affiliation(s)
- Kate Milward
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
59
|
|
60
|
Optimizing regulatory T cells for therapeutic application in human organ transplantation. Curr Opin Organ Transplant 2018; 23:516-523. [DOI: 10.1097/mot.0000000000000561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
61
|
European Reflections on New Indications for Extracorporeal Photopheresis in Solid Organ Transplantation. Transplantation 2018; 102:1279-1283. [DOI: 10.1097/tp.0000000000002244] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
62
|
Landman S, de Oliveira VL, van Erp PEJ, Fasse E, Bauland SCG, Joosten I, Koenen HJPM. Intradermal injection of low dose human regulatory T cells inhibits skin inflammation in a humanized mouse model. Sci Rep 2018; 8:10044. [PMID: 29968819 PMCID: PMC6030170 DOI: 10.1038/s41598-018-28346-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 06/11/2018] [Indexed: 12/31/2022] Open
Abstract
Recent regulatory T cell (Treg) based clinical trials support their therapeutic potential in transplantation and auto-inflammatory diseases. However, large numbers of Treg are needed to accomplish therapeutic efficacy. Local injection at the site of inflammation (targeted delivery) may lower the numbers needed for therapy. We evaluated if local delivery of low numbers of human Treg by intradermal injection was able to prevent skin inflammation, using the humanized mouse huPBL-SCID-huSkin allograft model. A dose of only 1 × 105 freshly isolated, non expanded Treg injected intradermally in close proximity to the transplanted human skin prevented inflammation of the grafted tissue induced by 4 × 107 IP injected human allogeneic PBMCs, (ratio Treg:PBMC = 1:400), as indicated by the inhibition of epidermal thickening, sustained Keratin-10 expression, the absence of Keratin-16 up regulation and prevention of human CD3+ T cell influx. A concomitant reduction of human T cells was observed in lymph nodes and spleen of the mice. Injection of Treg at the contralateral side was also shown to inhibit skin inflammation, suggesting that the inflammatory response was regulated both locally and systemically. In conclusion, local application of Treg may be an attractive way to suppress inflammation in vivo without the need for prior ex vivo expansion.
Collapse
Affiliation(s)
- Sija Landman
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Vivian L de Oliveira
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Piet E J van Erp
- Radboud university medical center, department of Dermatology, Nijmegen, The Netherlands
| | - Esther Fasse
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | | | - Irma Joosten
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands.
| |
Collapse
|
63
|
Ezzelarab MB. Regulatory T cells from allo- to xenotransplantation: Opportunities and challenges. Xenotransplantation 2018; 25:e12415. [DOI: 10.1111/xen.12415] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Mohamed B. Ezzelarab
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh Medical Center; Pittsburgh PA USA
| |
Collapse
|
64
|
Kawai K, Uchiyama M, Hester J, Wood K, Issa F. Regulatory T cells for tolerance. Hum Immunol 2018; 79:294-303. [DOI: 10.1016/j.humimm.2017.12.013] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/16/2017] [Accepted: 12/26/2017] [Indexed: 12/29/2022]
|
65
|
Abstract
CD4+CD25highFoxP3+ T regulatory cells (Tregs) are immunodominant suppressors in the immune system. Tregs use various mechanisms to control immune responses. Preclinical data from animal models have confirmed the huge therapeutic potential of Tregs in many immune-mediated diseases. Hence, these cells are now on the road to translation to cell therapy in the clinic as the first clinical trials are accomplished. To date, clinical research has involved mainly hematopoietic stem cell transplantations, solid organ transplantations, and autoimmunity. Despite difficulties with legislation and technical issues, treatment is constantly evolving and may soon represent a valid alternative for patients with diseases that are currently incurable. This review focuses on the basic and clinical experience with Tregs with adoptive transfer of these cells, primarily from clinical trials, as well as on perspectives on clinical use and technical problems with implementing the therapy.
Collapse
|
66
|
Garg G, Nikolouli E, Hardtke-Wolenski M, Toker A, Ohkura N, Beckstette M, Miyao T, Geffers R, Floess S, Gerdes N, Lutgens E, Osterloh A, Hori S, Sakaguchi S, Jaeckel E, Huehn J. Unique properties of thymic antigen-presenting cells promote epigenetic imprinting of alloantigen-specific regulatory T cells. Oncotarget 2018; 8:35542-35557. [PMID: 28415767 PMCID: PMC5482597 DOI: 10.18632/oncotarget.16221] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/03/2017] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) are potential immunotherapeutic candidates to induce transplantation tolerance. However, stability of Tregs still remains contentious and may potentially restrict their clinical use. Recent work suggested that epigenetic imprinting of Foxp3 and other Treg-specific signature genes is crucial for stabilization of immunosuppressive properties of Foxp3+ Tregs, and that these events are initiated already during early stages of thymic Treg development. However, the mechanisms governing this process remain largely unknown. Here we demonstrate that thymic antigen-presenting cells (APCs), including thymic dendritic cells (t-DCs) and medullary thymic epithelial cells (mTECs), can induce a more pronounced demethylation of Foxp3 and other Treg-specific epigenetic signature genes in developing Tregs when compared to splenic DCs (sp-DCs). Transcriptomic profiling of APCs revealed differential expression of secreted factors and costimulatory molecules, however neither addition of conditioned media nor interference with costimulatory signals affected Foxp3 induction by thymic APCs in vitro. Importantly, when tested in vivo both mTEC- and t-DC-generated alloantigen-specific Tregs displayed significantly higher efficacy in prolonging skin allograft acceptance when compared to Tregs generated by sp-DCs. Our results draw attention to unique properties of thymic APCs in initiating commitment towards stable and functional Tregs, a finding that could be highly beneficial in clinical immunotherapy.
Collapse
Affiliation(s)
- Garima Garg
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Eirini Nikolouli
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Matthias Hardtke-Wolenski
- Department of Gastroenterology, Hepatology, Endocrinology, Hannover Medical School, Hannover, Germany
| | - Aras Toker
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Naganari Ohkura
- Department of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Michael Beckstette
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Takahisa Miyao
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Floess
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Norbert Gerdes
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany.,Division of Cardiology, Pulmonology, and Vascular Medicine Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Esther Lutgens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany.,Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, AZ, Amsterdam, The Netherlands
| | - Anke Osterloh
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Shohei Hori
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan
| | - Shimon Sakaguchi
- Department of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology, Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
67
|
Duggleby R, Danby RD, Madrigal JA, Saudemont A. Clinical Grade Regulatory CD4 + T Cells (Tregs): Moving Toward Cellular-Based Immunomodulatory Therapies. Front Immunol 2018; 9:252. [PMID: 29487602 PMCID: PMC5816789 DOI: 10.3389/fimmu.2018.00252] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/29/2018] [Indexed: 12/26/2022] Open
Abstract
Regulatory T cells (Tregs) are CD4+ T cells that are key players of immune tolerance. They are powerful suppressor cells, able to impact the function of numerous immune cells, including key effectors of inflammation such as effector T cells. For this reason, Tregs are an ideal candidate for the development of cell therapy approaches to modulate immune responses. Treg therapy has shown promising results so far, providing key knowledge on the conditions in which these cells can provide protection and demonstrating that they could be an alternative to current pharmacological immunosuppressive therapies. However, a more comprehensive understanding of their characteristics, isolation, activation, and expansion is needed to be able design cost effective therapies. Here, we review the practicalities of making Tregs a viable cell therapy, in particular, discussing the challenges faced in isolating and manufacturing Tregs and defining what are the most appropriate applications for this new therapy.
Collapse
Affiliation(s)
- Richard Duggleby
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| | - Robert David Danby
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom.,Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| | - Aurore Saudemont
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| |
Collapse
|
68
|
Fraser H, Safinia N, Grageda N, Thirkell S, Lowe K, Fry LJ, Scottá C, Hope A, Fisher C, Hilton R, Game D, Harden P, Bushell A, Wood K, Lechler RI, Lombardi G. A Rapamycin-Based GMP-Compatible Process for the Isolation and Expansion of Regulatory T Cells for Clinical Trials. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 8:198-209. [PMID: 29552576 PMCID: PMC5850906 DOI: 10.1016/j.omtm.2018.01.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/16/2018] [Indexed: 01/09/2023]
Abstract
The concept of regulatory T cell (Treg)-based immunotherapy has enormous potential for facilitating tolerance in autoimmunity and transplantation. Clinical translation of Treg cell therapy requires production processes that satisfy the rigors of Good Manufacturing Practice (GMP) standards. In this regard, we report our findings on the implementation of a robust GMP compliant process for the ex vivo expansion of clinical grade Tregs, demonstrating the feasibility of this developed process for the manufacture of a final product for clinical application. This Treg isolation procedure ensured the selection of a pure Treg population that underwent a 300-fold expansion after 36 days of culture, while maintaining a purity of more than 75% CD4+CD25+FOXP3+ cells and a suppressive function of above 80%. Furthermore, we report the successful cryopreservation of the final product, demonstrating the maintenance of phenotype and function. The process outlined in this manuscript has been implemented in the ONE study, a multicenter phase I/IIa clinical trial in which cellular therapy is investigated in renal transplantation.
Collapse
Affiliation(s)
- Henrieta Fraser
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Niloufar Safinia
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Nathali Grageda
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Sarah Thirkell
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Katie Lowe
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Laura J Fry
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Cristiano Scottá
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Andrew Hope
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Christopher Fisher
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Rachel Hilton
- The Department of Nephrology and Transplantation, Guy's Hospital, Guy's and St. Thomas NHS Foundation Trust
| | - David Game
- The Department of Nephrology and Transplantation, Guy's Hospital, Guy's and St. Thomas NHS Foundation Trust
| | | | - Andrew Bushell
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Kathryn Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Robert I Lechler
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Giovanna Lombardi
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| |
Collapse
|
69
|
|
70
|
Zhu P, Atkinson C, Dixit S, Cheng Q, Tran D, Patel K, Jiang YL, Esckilsen S, Miller K, Bazzle G, Allen P, Moore A, Broome AM, Nadig SN. Organ preservation with targeted rapamycin nanoparticles: a pre-treatment strategy preventing chronic rejection in vivo. RSC Adv 2018; 8:25909-25919. [PMID: 30220998 PMCID: PMC6124302 DOI: 10.1039/c8ra01555d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/24/2018] [Indexed: 12/20/2022] Open
Abstract
Hypothermic preservation is the standard of care for storing organs prior to transplantation. Endothelial and epithelial injury associated with hypothermic storage causes downstream graft injury and, as such, the choice of an ideal donor organ preservation solution remains controversial. Cold storage solutions, by design, minimize cellular necrosis and optimize cellular osmotic potential, but do little to assuage immunological cell activation or immune cell priming post transplantation. Thus, here we explore the efficacy of our previously described novel Targeted Rapamycin Micelles (TRaM) as an additive to standard-of-care University of Wisconsin preservation solution as a means to alter the immunological microenvironment post transplantation using in vivo models of tracheal and aortic allograft transplantation. In all models of transplantation, grafts pre-treated with 100 ng mL-1 of TRaM augmented preservation solution ex vivo showed a significant inhibition of chronic rejection post-transplantation, as compared to UW augmented with free rapamycin at a ten-fold higher dose. Here, for the first time, we present a novel method of organ pretreatment using a nanotherapeutic-based cellular targeted delivery system that enables donor administration of rapamycin, at a ten-fold decreased dose during cold storage. Clinically, these pretreatment strategies may positively impact post-transplant outcomes and can be readily translated to clinical scenarios.
Collapse
Affiliation(s)
- Peng Zhu
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA.,Institute of Organ Transplantation, Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Carl Atkinson
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Suraj Dixit
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA.
| | - Qi Cheng
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA.,Institute of Organ Transplantation, Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danh Tran
- Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Kunal Patel
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Yu-Lin Jiang
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA.
| | - Scott Esckilsen
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Kayla Miller
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA.
| | - Grace Bazzle
- Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Patterson Allen
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Alfred Moore
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA.
| | - Ann-Marie Broome
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA. .,Department of Bioengineering, Clemson University, USA
| | - Satish N Nadig
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| |
Collapse
|
71
|
Schlöder J, Berges C, Tuettenberg A, Jonuleit H. Novel Concept of CD4-Mediated Activation of Regulatory T Cells for the Treatment of Graft-Versus-Host Disease. Front Immunol 2017; 8:1495. [PMID: 29167672 PMCID: PMC5682297 DOI: 10.3389/fimmu.2017.01495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/24/2017] [Indexed: 01/28/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is the only curative treatment option for several hematological malignancies and immune deficiency syndromes. Nevertheless, the development of a graft-versus-host disease (GvHD) after transplantation is a high risk and a severe complication with high morbidity and mortality causing therapeutic challenges. Current pharmacological therapies of GvHD lead to generalized immunosuppression followed by severe adverse side effects including infections and relapse of leukemia. Several novel cell-based immunomodulatory strategies for treatment or prevention of GvHD have been developed. Herein, thymus-derived regulatory T cells (tTreg), essential for the maintenance of peripheral immunologic tolerance, are in the focus of investigation. However, due to the limited number of tTreg in the peripheral blood, a complex, time- and cost-intensive in vitro expansion protocol is necessary for the production of an efficient cellular therapeutic. We demonstrated that activation of tTreg using the CD4-binding human immunodeficiency virus-1 protein gp120 leads to a substantially increased suppressor activity of tTreg without the need for additional expansion. Gp120-activated tTreg prevent GvHD development in a preclinical humanized mouse model. In addition, gp120 is not only effective in prevention but also in therapy of GvHD by suppressing all clinical symptoms and improving survival of treated mice. These data indicate that tTreg activation by gp120 is a feasible and potent strategy for significant functional improvement of tTreg as cellular therapeutic for GvHD treatment without the need of complicated, time-intensive, and expensive in vitro expansion of isolated tTreg.
Collapse
Affiliation(s)
- Janine Schlöder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Carsten Berges
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Helmut Jonuleit
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
72
|
Pilat N, Granofszky N, Wekerle T. Combining Adoptive Treg Transfer with Bone Marrow Transplantation for Transplantation Tolerance. CURRENT TRANSPLANTATION REPORTS 2017; 4:253-261. [PMID: 29201599 PMCID: PMC5691126 DOI: 10.1007/s40472-017-0164-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The mixed chimerism approach is an exceptionally potent strategy for the induction of donor-specific tolerance in organ transplantation and so far the only one that was demonstrated to work in the clinical setting. Regulatory T cells (Tregs) have been shown to improve chimerism induction in experimental animal models. This review summarizes the development of innovative BMT protocols using therapeutic Treg transfer for tolerance induction. RECENT FINDINGS Treg cell therapy promotes BM engraftment in reduced conditioning protocols in both, mice and non-human primates. In mice, transfer of polyclonal recipient Tregs was sufficient to substitute cytotoxic recipient conditioning. Treg therapy prevented chronic rejection of skin and heart allografts related to tissue-specific antigen disparities, in part by promoting intragraft Treg accumulation. SUMMARY Adoptive Treg transfer is remarkably effective in facilitating BM engraftment in reduced-intensity protocols in mice and non-human primates. Furthermore, it promotes regulatory mechanisms that prevent chronic rejection.
Collapse
Affiliation(s)
- Nina Pilat
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Nicolas Granofszky
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Thomas Wekerle
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
73
|
Madsen JC. Advances in the immunology of heart transplantation. J Heart Lung Transplant 2017; 36:1299-1305. [PMID: 29173391 DOI: 10.1016/j.healun.2017.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/16/2017] [Indexed: 12/30/2022] Open
Affiliation(s)
- Joren C Madsen
- Center for Transplantation Sciences and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
74
|
Liao T, Xue Y, Zhao D, Li S, Liu M, Chen J, Brand DD, Zheng H, Zhang Y, Zheng SG, Sun Q. In Vivo Attenuation of Antibody-Mediated Acute Renal Allograft Rejection by Ex Vivo TGF-β-Induced CD4 +Foxp3 + Regulatory T Cells. Front Immunol 2017; 8:1334. [PMID: 29085374 PMCID: PMC5650643 DOI: 10.3389/fimmu.2017.01334] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/02/2017] [Indexed: 11/28/2022] Open
Abstract
Antibody-mediated rejection (AMR) has emerged as the major cause of renal allograft dysfunction, and more effective strategies need to be explored for improving transplant outcomes. Regulatory T cells (Tregs), consisting of at least natural and induced Treg subsets, suppress effector responses at multiple levels and play a key role in transplantation tolerance. In this study, we investigated the effect of induced Tregs (iTregs) on preventing antibody-mediated renal injury and rejection in a mouse model. We observed that infusion of iTregs markedly attenuated histological graft injury and rejection and significantly improved renal allograft survival. iTregs exhibited a comprehensive ability to regulate immunological disorders in AMR. First, iTreg treatment decreased the levels of circulating antidonor antibody and the antibody deposition within allografts. Second, iTregs significantly reduced cell infiltration including CD4+ T cells (including Th1, Th17, and Tfh), CD8+IFN-γ+ cells, natural killer cells, B cells, and plasma cells, which are involved in the process of AMR. Our results also highlight a predominance of M1 macrophage infiltration in grafts with acute AMR, and M1 macrophage could be reduced by iTreg treatment. Collectively, our data demonstrate, for the first time, that TGF-β-induced Tregs can attenuate antibody-mediated acute renal allograft injury through targeting multiple effectors. Thus, use of iTregs in prevention of AMR in clinical practice could be expected.
Collapse
Affiliation(s)
- Tao Liao
- Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Youqiu Xue
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| | - Daqiang Zhao
- Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Siwen Li
- Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mingyu Liu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Science of Sun Yat-sen University, Guangzhou, China
| | - Jingrong Chen
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Haofeng Zheng
- Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yannan Zhang
- Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Song Guo Zheng
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| | - Qiquan Sun
- Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
75
|
Zaitsu M, Issa F, Hester J, Vanhove B, Wood KJ. Selective blockade of CD28 on human T cells facilitates regulation of alloimmune responses. JCI Insight 2017; 2:89381. [PMID: 28978798 DOI: 10.1172/jci.insight.89381] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/29/2017] [Indexed: 01/05/2023] Open
Abstract
T cells are central to the detrimental alloresponses that develop in autoimmunity and transplantation, with CD28 costimulatory signals being key to T cell activation and proliferation. CTLA4-Ig molecules that bind CD80/86 and inhibit CD28 costimulation offer an alternative immunosuppressive treatment, free from some of the chronic toxicities associated with calcineurin inhibition. However, CD80/86 blockade by CTLA4-Ig also results in the loss of coinhibitory CTLA4 signals that are critical to the regulation of T cell activation. Here, we show that a nonactivating monovalent anti-CD28 that spares CTLA4 signaling is an effective immunosuppressant in a clinically relevant humanized mouse transplant model. We demonstrate that selective CD28 blockade prolongs human skin allograft survival through a mechanism that includes a reduction in the cellular graft infiltrate. Critically, selective CD28 blockade promotes Treg function in vivo and synergizes with adoptive Treg therapy to promote transplant survival. In contrast to CTLA4-Ig treatment, selective CD28 blockade promotes regulation of alloimmune responses and facilitates Treg-based cellular therapy.
Collapse
Affiliation(s)
- Masaaki Zaitsu
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Bernard Vanhove
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| |
Collapse
|
76
|
Milward KF, Wood KJ, Hester J. Enhancing human regulatory T cells in vitro for cell therapy applications. Immunol Lett 2017; 190:139-147. [DOI: 10.1016/j.imlet.2017.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 12/25/2022]
|
77
|
Lee JS. Immunologic Mechanism of Ischemia Reperfusion Injury in Transplantation. KOREAN JOURNAL OF TRANSPLANTATION 2017. [DOI: 10.4285/jkstn.2017.31.3.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Jong Soo Lee
- Division of Nephrology, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
- Biomedical Research Center, Ulsan, Korea
| |
Collapse
|
78
|
Pilat N, Wekerle T. Combining Treg therapy with mixed chimerism: Getting the best of both worlds. CHIMERISM 2017; 1:26-9. [PMID: 21327149 DOI: 10.4161/chim.1.1.12964] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 07/12/2010] [Indexed: 11/19/2022]
Abstract
Deliberate establishment of donor-specific immunologic tolerance is considered to be the "Holy Grail" in transplantation medicine, but clinical tolerance protocols for routine organ transplantation are still an unmet need. Mixed hematopoietic chimerism is an attractive tolerance strategy with considerable potential. Recent pilot trials provide proof-of-principle that mixed chimerism can induce tolerance in renal transplant recipients. Routine clinical translation, however, is impeded by the side effects of the cytotoxic recipient conditioning necessary for the transient engraftment of HLA-mismatched BM. In murine studies recently published in The American Journal of Transplantation, we demonstrated that the therapeutic application of polyclonal recipient regulatory T cells (Tregs) leads to engraftment of practicable doses of fully allogeneic BM and to donor-specific tolerance without any cytotoxic conditioning, thereby eliminating a major impediment for the clinical translation of the mixed chimerism strategy in the experimental setting. The background and the implications of these findings are discussed.
Collapse
Affiliation(s)
- Nina Pilat
- Division of Transplantation; Department of Surgery; Vienna General Hospital; Medical University of Vienna; Vienna, Austria
| | | |
Collapse
|
79
|
Histone/protein deacetylase 11 targeting promotes Foxp3+ Treg function. Sci Rep 2017; 7:8626. [PMID: 28819166 PMCID: PMC5561267 DOI: 10.1038/s41598-017-09211-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/21/2017] [Indexed: 12/13/2022] Open
Abstract
Current interest in Foxp3+ T-regulatory (Treg) cells as therapeutic targets in transplantation is largely focused on their harvesting pre-transplant, expansion and infusion post-transplantation. An alternate strategy of pharmacologic modulation of Treg function using histone/protein deacetylase inhibitors (HDACi) may allow more titratable and longer-term dosing. However, the effects of broadly acting HDACi vary, such that HDAC isoform-selective targeting is likely required. We report data from mice with constitutive or conditional deletion of HDAC11 within Foxp3+ Treg cells, and their use, along with small molecule HDAC11 inhibitors, in allograft models. Global HDAC11 deletion had no effect on health or development, and compared to WT controls, Foxp3+ Tregs lacking HDAC11 showed increased suppressive function, and increased expression of Foxp3 and TGF-β. Likewise, compared to WT recipients, conditional deletion of HDAC11 within Tregs led to long-term survival of fully MHC-mismatched cardiac allografts, and prevented development of transplant arteriosclerosis in an MHC class II-mismatched allograft model. The translational significance of HDAC11 targeting was shown by the ability of an HDAC11i to promote long-term allograft allografts in fully MHC-disparate strains. These data are powerful stimuli for the further development and testing of HDAC11-selective pharmacologic inhibitors, and may ultimately provide new therapies for transplantation and autoimmune diseases.
Collapse
|
80
|
Kang N, Toyofuku WM, Yang X, Scott MD. Inhibition of allogeneic cytotoxic T cell (CD8 +) proliferation via polymer-induced Treg (CD4 +) cells. Acta Biomater 2017; 57:146-155. [PMID: 28442414 DOI: 10.1016/j.actbio.2017.04.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/13/2017] [Accepted: 04/21/2017] [Indexed: 01/16/2023]
Abstract
T cell-mediated immune rejection remains a barrier to successful transplantation. Polymer-based bioengineering of cells may provide an effective means of preventing allorecognition and the proliferation of cytotoxic (CD8+) T lymphocytes (CTL). Using MHC-disparate murine splenocytes modified with succinimidyl valerate activated methoxypoly(ethylene glycol) [SVA-mPEG] polymers, the effects of leukocyte immunocamouflage on CD8+ and CD4+ alloproliferation and T regulatory (Treg) cell induction were assessed in a mixed lymphocyte reaction (MLR) model. Polymer-grafting effectively camouflaged multiple leukocyte markers (MHC class I and II, TCR and CD3) essential for effective allorecognition. Consequent to the polymer-induced immunocamouflage of the cell membrane, both CD8+ and CD4+ T cell alloproliferation were significantly inhibited in a polymer dose-dependent manner. The loss of alloproliferation correlated with the induction of Treg cells (CD4+CD25+Foxp3+). The Tregs, surprisingly, arose primarily via differentiation of naive, non-proliferating, CD4+ cells. Of biologic importance, the polymer-induced Treg were functional and exhibited potent immunosuppressive activity on allogeneic CTL proliferation. These results suggest that immunocamouflage-mediated attenuation of alloantigen-TCR recognition can prevent the tissue destructive allogeneic CD8+ T cell response, both directly and indirectly, through the generation/differentiation of functional Tregs. Immunocamouflage induced tolerance could be clinically valuable in attenuating T cell-mediated transplant rejection and in the treatment of autoimmune diseases. STATEMENT OF SIGNIFICANCE While our previous studies have demonstrated that polymer-grafting to MHC disparate leukocytes inhibits CD4+ cell proliferation, the effects of PEGylation on the alloproliferation of CD8+ cytotoxic T cells (CTL) was not examined. As shown here, PEGylation of allogeneic leukocytes prevents the generation of the CTL response responsible for acute rejection. The loss of CTL proliferation is consequent to the polymer-based attenuation of allorecognition and the induction of T regulatory cells (Tregs). Interestingly, the Tregs are primarily generated via the differentiation of non-proliferating naive T cells. Importantly, the Tregs are functional and effectively induce a tolerogenic environment when transferred to an alloresponsive environment. The use of polymer-modified leukocytes provides a unique approach to effectively maximize the biologic production of functional Tregs both in vitro and in vivo. By using this approach it may be possible to attenuate unwanted alloresponses (e.g., graft rejection) or to treat autoimmune diseases.
Collapse
Affiliation(s)
- Ning Kang
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Wendy M Toyofuku
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Xining Yang
- University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Mark D Scott
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
81
|
Nikolouli E, Hardtke-Wolenski M, Hapke M, Beckstette M, Geffers R, Floess S, Jaeckel E, Huehn J. Alloantigen-Induced Regulatory T Cells Generated in Presence of Vitamin C Display Enhanced Stability of Foxp3 Expression and Promote Skin Allograft Acceptance. Front Immunol 2017; 8:748. [PMID: 28702031 PMCID: PMC5487376 DOI: 10.3389/fimmu.2017.00748] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
Regulatory T cells (Tregs) are critical for the maintenance of immune homeostasis and self-tolerance and can be therapeutically used for prevention of unwanted immune responses such as allotransplant rejection. Tregs are characterized by expression of the transcription factor Foxp3, and recent work suggests that epigenetic imprinting of Foxp3 and other Treg-specific epigenetic signatures genes is crucial for the stabilization of both Foxp3 expression and immunosuppressive properties within Tregs. Lately, vitamin C was reported to enhance the activity of enzymes of the ten-eleven translocation family, thereby fostering the demethylation of Foxp3 and other Treg-specific epigenetic signatures genes in developing Tregs. Here, we in vitro generated alloantigen-induced Foxp3+ Tregs (allo-iTregs) in presence of vitamin C. Although vitamin C hardly influenced the transcriptome of allo-iTregs as revealed by RNA-seq, those vitamin C-treated allo-iTregs showed a more pronounced demethylation of Foxp3 and other Treg-specific epigenetic signatures genes accompanied with an enhanced stability of Foxp3 expression. Accordingly, when being tested in vivo in an allogeneic skin transplantation model, vitamin C-treated allo-iTregs showed a superior suppressive capacity. Together, our results pave the way for the establishment of novel protocols for the in vitro generation of alloantigen-induced Foxp3+ Tregs for therapeutic use in transplantation medicine.
Collapse
Affiliation(s)
- Eirini Nikolouli
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Martin Hapke
- Department Gastroenterology, Hepatology, Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael Beckstette
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Elmar Jaeckel
- Department Gastroenterology, Hepatology, Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
82
|
Innate Immune Response in Kidney Ischemia/Reperfusion Injury: Potential Target for Therapy. J Immunol Res 2017; 2017:6305439. [PMID: 28676864 PMCID: PMC5476886 DOI: 10.1155/2017/6305439] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/17/2017] [Indexed: 01/06/2023] Open
Abstract
Acute kidney injury caused by ischemia and subsequent reperfusion is associated with a high rate of mortality and morbidity. Ischemia/reperfusion injury in kidney transplantation causes delayed graft function and is associated with more frequent episodes of acute rejection and progression to chronic allograft nephropathy. Alloantigen-independent inflammation is an important process, participating in pathogenesis of injurious response, caused by ischemia and reperfusion. This innate immune response is characterized by the activity of classical cells belonging to the immune system, such as neutrophils, macrophages, dendritic cells, lymphocytes, and also tubular epithelial cells and endothelial cells. These immune cells not only participate in inflammation after ischemia exerting detrimental influence but also play a protective role in the healing response from ischemia/reperfusion injury. Delineating of complex mechanisms of their actions could be fruitful in future prevention and treatment of ischemia/reperfusion injury. Among numerous so far conducted experiments, observed immunomodulatory role of adenosine and adenosine receptor agonists in complex interactions of dendritic cells, natural killer T cells, and T regulatory cells is emphasized as promising in the treatment of kidney ischemia/reperfusion injury. Potential pharmacological approaches which decrease NF-κB activity and antagonize mechanisms downstream of activated Toll-like receptors are discussed.
Collapse
|
83
|
Salman J, Ius F, Knoefel AK, Sommer W, Siemeni T, Kuehn C, Tudorache I, Avsar M, Nakagiri T, Preissler G, Hatz R, Greer M, Welte T, Haverich A, Warnecke G. Association of Higher CD4 + CD25 high CD127 low , FoxP3 + , and IL-2 + T Cell Frequencies Early After Lung Transplantation With Less Chronic Lung Allograft Dysfunction at Two Years. Am J Transplant 2017; 17:1637-1648. [PMID: 27931084 DOI: 10.1111/ajt.14148] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 11/13/2016] [Accepted: 11/25/2016] [Indexed: 01/25/2023]
Abstract
Regulatory T cells (Treg) can regulate alloantigens and may counteract chronic lung allograft dysfunction (CLAD) in lung transplantation. We analyzed Treg in peripheral blood prospectively and correlated percentages of subpopulations with the incidence of CLAD at 2 years. Among lung-transplanted patients between January 2009 and July 2011, only patients with sufficient Treg measurements were included into the study. Tregs were measured immediately before lung transplantation, at 3 weeks and 3, 6, 12, and 24 months after transplantation and were defined as CD4+ CD25high T cells and further analyzed for CTLA4, CD127, FoxP3, and IL-2 expressions. Between January 2009 and July 2011, 264 patients were transplanted at our institution. Among the 138 (52%) patients included into the study, 31 (22%) developed CLAD within 2 years after transplantation. As soon as 3 weeks after lung transplantation, a statistically significant positive association was detected between Treg frequencies and later absence of CLAD. At the multivariate analysis, increasing frequencies of CD4+ CD25high CD127low , CD4+ CD25high FoxP3+ and CD4+ CD25high IL-2+ T cells at 3 weeks after lung transplantation emerged as protective factors against development of CLAD at 2 years. In conclusion, higher frequencies of specific Treg subpopulations early after lung transplantation are protective against CLAD development.
Collapse
Affiliation(s)
- J Salman
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - F Ius
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - A-K Knoefel
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - W Sommer
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - T Siemeni
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - C Kuehn
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - I Tudorache
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - M Avsar
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - T Nakagiri
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - G Preissler
- Department of Surgery, Munich Lung Transplant Group, Ludwig-Maximilian's University, Munich, Germany
| | - R Hatz
- Department of Surgery, Munich Lung Transplant Group, Ludwig-Maximilian's University, Munich, Germany
| | - M Greer
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - T Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - A Haverich
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - G Warnecke
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
84
|
Vaikunthanathan T, Safinia N, Boardman D, Lechler RI, Lombardi G. Regulatory T cells: tolerance induction in solid organ transplantation. Clin Exp Immunol 2017; 189:197-210. [PMID: 28422316 DOI: 10.1111/cei.12978] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
The concept of regulatory T cell (Treg ) therapy in transplantation is now a reality. Significant advances in science and technology have enabled us to isolate human Tregs , expand them to clinically relevant numbers and infuse them into human transplant recipients. With several Phase I/II trials under way investigating Treg safety and efficacy it is now more crucial than ever to understand their complex biology. However, our journey is by no means complete; results from these trials will undoubtedly provoke both further knowledge and enquiry which, alongside evolving science, will continue to drive the optimization of Treg therapy in the pursuit of transplantation tolerance. In this review we will summarize current knowledge of Treg biology, explore novel technologies in the setting of Treg immunotherapy and address key prerequisites surrounding the clinical application of Tregs in transplantation.
Collapse
Affiliation(s)
- T Vaikunthanathan
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - N Safinia
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - D Boardman
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - R I Lechler
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - G Lombardi
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| |
Collapse
|
85
|
A20 Haploinsufficiency Aggravates Transplant Arteriosclerosis in Mouse Vascular Allografts: Implications for Clinical Transplantation. Transplantation 2017; 100:e106-e116. [PMID: 27495763 DOI: 10.1097/tp.0000000000001407] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Inflammation is central to the pathogenesis of transplant arteriosclerosis (TA). We questioned whether physiologic levels of anti-inflammatory A20 influence TA severity. METHODS We performed major histocompatibility complex mismatched aorta to carotid artery interposition grafts, using wild type (WT) or A20 heterozygote (HET) C57BL/6 (H-2) donors and BALB/c (H-2) recipients, and conversely BALB/c donors and WT/HET recipients. We analyzed aortic allografts by histology, immunohistochemistry, immunofluorescence, and gene profiling (quantitative real-time reverse-transcriptase polymerase chain reaction). We validated select in vivo A20 targets in human and mouse smooth muscle cell (SMC) cultures. RESULTS We noted significantly greater intimal hyperplasia in HET versus WT allografts, indicating aggravated TA. Inadequate upregulation of A20 in HET allografts after transplantation was associated with excessive NF-кB activation, gauged by higher levels of IkBα, p65, VCAM-1, ICAM-1, CXCL10, CCL2, TNF, and IL-6 (mostly localized to SMC). Correspondingly, cytokine-induced upregulation of TNF and IL-6 in human and mouse SMC cultures inversely correlated with A20 expression. Aggravated TA in HET versus WT allografts correlated with increased intimal SMC proliferation, and a higher number of infiltrating IFNγ and Granzyme B CD4 T cells and natural killer cells, and lower number of FoxP3 regulatory T cells. A20 haploinsufficiency in allograft recipients did not influence TA. CONCLUSIONS A20 haploinsufficiency in vascular allografts aggravates lesions of TA by exacerbating inflammation, SMC proliferation, and infiltration of pathogenic T cells. A20 single nucleotide polymorphisms associating with lower A20 expression or function in donors of vascularized allografts may inform risk and severity of TA, highlighting the clinical implications of our findings.
Collapse
|
86
|
Korczak-Kowalska G, Stelmaszczyk-Emmel A, Bocian K, Kiernozek E, Drela N, Domagała-Kulawik J. Expanding Diversity and Common Goal of Regulatory T and B Cells. II: In Allergy, Malignancy, and Transplantation. Arch Immunol Ther Exp (Warsz) 2017; 65:523-535. [PMID: 28470464 PMCID: PMC5688211 DOI: 10.1007/s00005-017-0471-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/19/2017] [Indexed: 01/03/2023]
Abstract
Regulation of immune response was found to play an important role in the course of many diseases such as autoimmune diseases, allergy, malignancy, organ transplantation. The studies on immune regulation focus on the role of regulatory cells (Tregs, Bregs, regulatory myeloid cells) in these disorders. The number and function of Tregs may serve as a marker of disease activity. As in allergy, the depletion of Tregs is observed and the results of allergen-specific immunotherapy could be measured by an increase in the population of IL-10+ regulatory cells. On the basis of the knowledge of anti-cancer immune response regulation, new directions in therapy of tumors are introduced. As the proportion of regulatory cells is increased in the course of neoplasm, the therapeutic action is directed at their inhibition. The depletion of Tregs may be also achieved by an anti-check-point blockade, anti-CD25 agents, and inhibition of regulatory cell recruitment to the tumor site by affecting chemokine pathways. However, the possible favorable role of Tregs in cancer development is considered and the plasticity of immune regulation should be taken into account. The new promising direction of the treatment based on regulatory cells is the prevention of transplant rejection. A different way of production and implementation of classic Tregs as well as other cell types such as double-negative cells, Bregs, CD4+ Tr1 cells are tested in ongoing trials. On the basis of the results of current studies, we could show in this review the significance of therapies based on regulatory cells in different disorders.
Collapse
Affiliation(s)
- Grażyna Korczak-Kowalska
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland
| | - Anna Stelmaszczyk-Emmel
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Bocian
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Ewelina Kiernozek
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Nadzieja Drela
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
87
|
Govender L, Wyss JC, Kumar R, Pascual M, Golshayan D. IL-2-Mediated In Vivo Expansion of Regulatory T Cells Combined with CD154-CD40 Co-Stimulation Blockade but Not CTLA-4 Ig Prolongs Allograft Survival in Naive and Sensitized Mice. Front Immunol 2017; 8:421. [PMID: 28484450 PMCID: PMC5399033 DOI: 10.3389/fimmu.2017.00421] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 03/24/2017] [Indexed: 01/03/2023] Open
Abstract
In recent years, regulatory T cells (Treg)-based immunotherapy has emerged as a promising strategy to promote operational tolerance after solid organ transplantation (SOT). However, a main hurdle for the therapeutic use of Treg in transplantation is their low frequency, particularly in non-lymphopenic hosts. We aimed to expand Treg directly in vivo and determine their efficacy in promoting donor-specific tolerance, using a stringent experimental model. Administration of the IL-2/JES6-1 immune complex at the time of transplantation resulted in significant expansion of donor-specific Treg, which suppressed alloreactive T cells. IL-2-mediated Treg expansion in combination with short-term CD154–CD40 co-stimulation blockade, but not CTLA-4 Ig or rapamycin, led to tolerance to MHC-mismatched skin grafts in non-lymphopenic mice, mainly by hindering alloreactive CD8+ effector T cells and the production of alloantibodies. Importantly, this treatment also allowed prolonged survival of allografts in the presence of either donor-specific or cross-reactive memory cells. However, late rejection occurred in sensitized hosts, partly mediated by activated B cells. Overall, these data illustrate the potential but also some important limitations of Treg-based therapy in clinical SOT as well as the importance of concomitant immunomodulatory strategies in particular in sensitized hosts.
Collapse
Affiliation(s)
- Lerisa Govender
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Jean-Christophe Wyss
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Rajesh Kumar
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Manuel Pascual
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
88
|
Patel K, Atkinson C, Tran D, Nadig SN. Nanotechnological Approaches to Immunosuppression and Tolerance Induction. CURRENT TRANSPLANTATION REPORTS 2017; 4:159-168. [PMID: 29057203 DOI: 10.1007/s40472-017-0146-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW Several preclinical studies have engineered nanoparticles for immune regulation, and have shown promising results in the fields of autoimmunity and cancer. In solid organ transplantation, the use of nanoparticle-based immune regulation has only just begun to emerge but holds significant promise for the improvement of our current standard of care immunosuppressive regimens. In this review, we will shed light on the current status of nanoparticle-engineered immunotherapeutics, and the potential application of these technologies to the field of organ transplantation. Further we discuss different strategies for delivery and potential cellular targeting moieties that could be utilized to obviate the need for high dose systemic immunosuppressive regimens. RECENT FINDINGS Recent studies have shown the potential of immunosuppressive laden nanoparticles to increase bioavailability, drug release, and specifically target immune cell compartments as methods to provide recipient immunosuppressive sparing strategies. SUMMARY Nanoparticle centered immunosuppressive strategies hold the potential to usher in a new era in transplant recipient management and could hold the key to minimizing off-target effects of immunosuppressants, along with prolonging transplant survival.
Collapse
Affiliation(s)
- Kunal Patel
- Department of Surgery, Division of Transplantation, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Division of Transplantation, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Carl Atkinson
- Department of Surgery, Division of Transplantation, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Division of Transplantation, Medical University of South Carolina, Charleston, South Carolina, USA
- South Carolina Investigators in Transplantation, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Danh Tran
- Department of Surgery, Division of Transplantation, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Division of Transplantation, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Satish N Nadig
- Department of Surgery, Division of Transplantation, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Division of Transplantation, Medical University of South Carolina, Charleston, South Carolina, USA
- South Carolina Investigators in Transplantation, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
89
|
Duguet F, Locard-Paulet M, Marcellin M, Chaoui K, Bernard I, Andreoletti O, Lesourne R, Burlet-Schiltz O, Gonzalez de Peredo A, Saoudi A. Proteomic Analysis of Regulatory T Cells Reveals the Importance of Themis1 in the Control of Their Suppressive Function. Mol Cell Proteomics 2017; 16:1416-1432. [PMID: 28373295 DOI: 10.1074/mcp.m116.062745] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 03/13/2017] [Indexed: 01/25/2023] Open
Abstract
Regulatory T cells (Treg) represent a minor subpopulation of T lymphocytes that is crucial for the maintenance of immune homeostasis. Here, we present a large-scale quantitative mass spectrometry study that defines a specific proteomic "signature" of Treg. Treg and conventional T lymphocyte (Tconv) subpopulations were sorted by flow cytometry and subjected to global proteomic analysis by single-run nanoLC-MS/MS on a fast-sequencing Q-Exactive mass spectrometer. Besides "historical" proteins that characterize Treg, our study identified numerous new proteins that are up- or downregulated in Treg versus Tconv. We focused on Themis1, a protein particularly under-represented in Treg, and recently described as being involved in the pathogenesis of immune diseases. Using a transgenic mouse model overexpressing Themis1, we provided in vivo and in vitro evidence of its importance for Treg suppressive functions, in an animal model of inflammatory bowel disease and in coculture assays. We showed that this enhanced suppressive activity in vitro is associated with an accumulation of Tregs. Thus, our study highlights the usefulness of label free quantitative methods to better characterize the Treg cell lineage and demonstrates the potential role of Themis1 in the suppressive functions of these cells.
Collapse
Affiliation(s)
- Fanny Duguet
- From the ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France.,§Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, UPS, 31024, Toulouse, France
| | - Marie Locard-Paulet
- From the ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Marlène Marcellin
- From the ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Karima Chaoui
- From the ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Isabelle Bernard
- §Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, UPS, 31024, Toulouse, France
| | - Olivier Andreoletti
- ¶UMR INRA ENVT 1225, Interactions Hôtes Agents Pathogènes, Ecole Nationale Vétérinaire de Toulouse, 31000 Toulouse, France
| | - Renaud Lesourne
- §Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, UPS, 31024, Toulouse, France
| | - Odile Burlet-Schiltz
- From the ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Anne Gonzalez de Peredo
- From the ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France;
| | - Abdelhadi Saoudi
- §Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, UPS, 31024, Toulouse, France
| |
Collapse
|
90
|
da Silva MB, da Cunha FF, Terra FF, Camara NOS. Old game, new players: Linking classical theories to new trends in transplant immunology. World J Transplant 2017; 7:1-25. [PMID: 28280691 PMCID: PMC5324024 DOI: 10.5500/wjt.v7.i1.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/16/2016] [Accepted: 12/07/2016] [Indexed: 02/05/2023] Open
Abstract
The evolutionary emergence of an efficient immune system has a fundamental role in our survival against pathogenic attacks. Nevertheless, this same protective mechanism may also establish a negative consequence in the setting of disorders such as autoimmunity and transplant rejection. In light of the latter, although research has long uncovered main concepts of allogeneic recognition, immune rejection is still the main obstacle to long-term graft survival. Therefore, in order to define effective therapies that prolong graft viability, it is essential that we understand the underlying mediators and mechanisms that participate in transplant rejection. This multifaceted process is characterized by diverse cellular and humoral participants with innate and adaptive functions that can determine the type of rejection or promote graft acceptance. Although a number of mediators of graft recognition have been described in traditional immunology, recent studies indicate that defining rigid roles for certain immune cells and factors may be more complicated than originally conceived. Current research has also targeted specific cells and drugs that regulate immune activation and induce tolerance. This review will give a broad view of the most recent understanding of the allogeneic inflammatory/tolerogenic response and current insights into cellular and drug therapies that modulate immune activation that may prove to be useful in the induction of tolerance in the clinical setting.
Collapse
|
91
|
Souidi N, Stolk M, Rudeck J, Strunk D, Schallmoser K, Volk HD, Seifert M. Stromal Cells Act as Guardians for Endothelial Progenitors by Reducing Their Immunogenicity After Co-Transplantation. Stem Cells 2017; 35:1233-1245. [PMID: 28100035 DOI: 10.1002/stem.2573] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/07/2016] [Accepted: 12/18/2016] [Indexed: 01/15/2023]
Abstract
Regeneration of injured tissues requires effective therapeutic strategies supporting vasculogenesis. The lack of instantly available autologous cell sources and immunogenicity of allogeneic endothelial (progenitor) cells limits clinical progress. Based on the immunosuppressive potency of mesenchymal stem/progenitor cells (MSCs), we investigated whether crosstalk between endothelial colony-forming progenitor cells (ECFCs) and MSCs during vasculogenesis could lower allogeneic T cell responses against ECFCs allowing long-term engraftment in vivo. Immunodeficient mice received subcutaneous grafts containing human ECFCs alone, or pairs of human ECFCs/MSCs from the same umbilical cord (UC) to study vasculogenesis in the presence of human leukocyte antigen (HLA)-mismatched human peripheral blood mononuclear cells (PBMCs). In vitro, cell surface marker changes due to interferon gamma (IFNγ) stimulation during ECFC/MSC coculture were determined and further effects on allostimulated T cell proliferation and cytotoxic lysis were measured. IFNγ-induced HLA-DR expression on ECFCs and MSCs, but both cell types had significantly less HLA-DR in cocultures. ECFC-induced T cell proliferation was abolished after MSC coculture as a result of HLA-DR downregulation and indolamin-2,3-dioxygenase activation. Additionally, allospecific CD8+ T cell-mediated lysis of ECFCs was reduced in cocultures. ECFC/MSC coapplication in immunodeficient mice not only promoted the generation of improved blood vessel architecture after 6 weeks, but also reduced intragraft immune cell infiltration and endothelial HLA-DR expression following PBMC reconstitution. Crosstalk between UC-derived ECFCs and MSCs after combined transplantation can lower the risk of ECFC rejection, thus enabling their coapplication for therapeutic vasculogenesis. Stem Cells 2017;35:1233-1245.
Collapse
Affiliation(s)
- Naima Souidi
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Meaghan Stolk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Juliane Rudeck
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Dirk Strunk
- Institute of Experimental and Clinical Cell Therapy, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord & Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Katharina Schallmoser
- Spinal Cord & Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria.,Department for Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Hans-Dieter Volk
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Martina Seifert
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| |
Collapse
|
92
|
Romano M, Tung SL, Smyth LA, Lombardi G. Treg therapy in transplantation: a general overview. Transpl Int 2017; 30:745-753. [PMID: 28012226 DOI: 10.1111/tri.12909] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/26/2016] [Accepted: 12/19/2016] [Indexed: 12/23/2022]
Abstract
Solid organ transplantation remains the treatment of choice for end-stage organ failure. Whilst the short-term outcomes post-transplant have improved in the last decades, chronic rejection and immunosuppressant side effects remain an ongoing concern. Hematopoietic stem cell transplantation is a well-established procedure for the treatment of patients with haematological disorders. However, donor T cells are continually primed and activated to react against the host causing graft-versus-host disease (GvHD) that leads to tissue damages and death. Regulatory T cells (Tregs) play an essential role in maintaining tolerance to self-antigens, preventing excessive immune responses and abrogating autoimmunity. Due to their suppressive properties, Tregs have been extensively studied for their use as a cellular therapy aiming to treat GvHD and limit immune responses responsible for graft rejection. Several clinical trials have been conducted or are currently ongoing to investigate safety and feasibility of Treg-based therapy. This review summarizes the general understanding of Treg biology and presents the methods used to isolate and expand Tregs. Furthermore, we describe data from the first clinical trials using Tregs, explaining the limitations and future application of these cells.
Collapse
Affiliation(s)
- Marco Romano
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Sim Lai Tung
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Lesley Ann Smyth
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,School of Health Sport and Bioscience, University of East London, London, UK
| | - Giovanna Lombardi
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
93
|
Arroyo Hornero R, Betts GJ, Sawitzki B, Vogt K, Harden PN, Wood KJ. CD45RA Distinguishes CD4+CD25+CD127-/low TSDR Demethylated Regulatory T Cell Subpopulations With Differential Stability and Susceptibility to Tacrolimus-Mediated Inhibition of Suppression. Transplantation 2017; 101:302-309. [PMID: 28118317 PMCID: PMC5265687 DOI: 10.1097/tp.0000000000001278] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/10/2016] [Accepted: 03/25/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Adoptive transfer of forkhead box protein (FOX)3 regulatory T (Treg) cells offers a promising strategy to reduce damage to an allograft by the recipient's immune system. Identification of cell surface markers sufficient to purify Treg cells expanded ex vivo to remove cellular contaminants requires optimization. Furthermore, the expanded Treg must be able to survive, expand, and suppress in allograft recipients exposed to immunosuppressants, such as tacrolimus (TAC). Reduced CD127 expression enhances identification of Treg in the human CD4CD25 population. CD45RA expression identifies naive CD4CD25 Treg with an enhanced stability of Treg phenotype. METHODS We combine an analysis of CD45RA, CD25, and CD127 expression to identify subpopulations of CD4CD127CD25 cells. Regulatory T cells were sorted according to expression of CD25 and CD45RA and expanded in the presence of a physiological relevant concentration of TAC. Regulatory T cell-specific demethylation region (TSDR) demethylation, FOXP3 expression, and suppression were analyzed. RESULTS CD4CD127CD25CD45RA Treg cells had a stable TSDR demethylated FOXP3 phenotype after expansion whereas CD4CD127CD25CD45RA Treg cell lost the TSDR demethylated phenotype. CD45RA Treg had a greater capacity to suppress after expansion with TAC. CONCLUSIONS Although CD45RA Treg retained a greater suppressive capacity when expanded with TAC, the marked loss of the TSDR demethylated status highlights the potential for loss of stability of these cells in transplant recipients treated with TAC based immunosuppression. We show that a population of CD4CD127CD45RA Regulatory T cell may offer the best compromise between susceptibility to loss of suppression when exposed to TAC and maintenance of a TSDR demethylated phenotype following in vitro expansion.
Collapse
Affiliation(s)
- Rebeca Arroyo Hornero
- 1 Nuffield Department of Surgical Sciences, Oxford University, John Radcliffe Hospital, Oxford, United Kingdom. 2 Charité-Universitätsmedizin Berlin, Campus Virchow, Institut f. Medizinische Immunologie, Berlin, Germany. 3 Oxford Transplant Centre, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | | | | | | | | | | |
Collapse
|
94
|
Ezzelarab MB, Thomson AW. Adoptive Cell Therapy with Tregs to Improve Transplant Outcomes: The Promise and the Stumbling Blocks. CURRENT TRANSPLANTATION REPORTS 2016; 3:265-274. [PMID: 28529840 PMCID: PMC5435383 DOI: 10.1007/s40472-016-0114-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The contribution of regulatory T cells (Treg) to the induction and maintenance of tolerance is well-recognized in rodents and may contribute to long-term human organ allograft survival. The therapeutic efficacy of adoptively-transferred Treg in promoting tolerance to organ allografts is well-recognized in mouse models. Early phase 1/2 clinical studies of Treg therapy have been conducted in patients with type-1 (autoimmune) diabetes and refractory Crohn's disease, and for inhibition of graft-versus-host disease following bone marrow transplantation with proven safety. The feasibility of adoptive Treg therapy in the clinic is subject to various parameters, including optimal cell source, isolation procedure, expansion, target dose, time of infusion, as well as generation of a GMP-cell product. Several phase 1/2 Treg dose-escalation studies are underway in organ transplantation. Recent evidence suggests that additional factors are critical to ensure Treg safety and efficacy in allograft recipients, including Treg characterization, stability, longevity, trafficking, concomitant immunosuppression, and donor antigen specificity. Accordingly, Treg therapy in the context of organ transplantation may prove more challenging in comparison to other prospective clinical settings of Treg immunotherapy, such as type-1 diabetes.
Collapse
Affiliation(s)
- Mohamed B. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Angus W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
95
|
Siemeni T, Knöfel AK, Madrahimov N, Sommer W, Avsar M, Salman J, Ius F, Frank N, Büchler G, Jonigk D, Jansson K, Maus U, Tudorache I, Falk CS, Haverich A, Warnecke G. In Vivo Development of Transplant Arteriosclerosis in Humanized Mice Reflects Alloantigen Recognition and Peripheral Treg Phenotype of Lung Transplant Recipients. Am J Transplant 2016; 16:3150-3162. [PMID: 27273729 DOI: 10.1111/ajt.13905] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 01/25/2023]
Abstract
Experimentally, regulatory T cells inhibit rejection. In clinical transplantations, however, it is not known whether T cell regulation is the cause for, or an epiphenomenon of, long-term allograft survival. Here, we study naïve and alloantigen-primed T cell responses of clinical lung transplant recipients in humanized mice. The pericardiophrenic artery procured from human lung grafts was implanted into the aorta of NODrag-/- /IL-2rγc-/- mice reconstituted with peripheral blood mononuclear cells (PBMCs) from the respective lung recipient. Naïve or primed allogeneic PBMCs procured 21 days post-lung transplantation with or without enriching for CD4+ CD25high T cells were used. Transplant arteriosclerosis was assessed 28 days later by histology. Mice reconstituted with alloantigen-primed PBMCs showed significantly more severe transplant arteriosclerosis than did mice with naïve PBMCs (p = 0.005). Transplant arteriosclerosis was equally suppressed by enriching for autologous naïve (p = 0.012) or alloantigen-primed regulatory T cells (Tregs) (p = 0.009). Alloantigen priming in clinical lung recipients can be adoptively transferred into a humanized mouse model. Transplant arteriosclerosis elicited by naïve or alloantigen-primed PBMCs can be similarly controlled by potent autologous Tregs. Cellular therapy with expanded autologous Tregs in lung transplantation might be a promising future strategy.
Collapse
Affiliation(s)
- T Siemeni
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - A-K Knöfel
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - N Madrahimov
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - W Sommer
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - M Avsar
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - J Salman
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - F Ius
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - N Frank
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - G Büchler
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - D Jonigk
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - K Jansson
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - U Maus
- German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany.,Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - I Tudorache
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - C S Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - A Haverich
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - G Warnecke
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany. .,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
96
|
Li L, Zhang Y, Xu M, Rong R, Wang J, Zhu T. Inhibition of histone methyltransferase EZH2 ameliorates early acute renal allograft rejection in rats. BMC Immunol 2016; 17:41. [PMID: 27784285 PMCID: PMC5080725 DOI: 10.1186/s12865-016-0179-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 10/17/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although histone methyltransferases EZH2 has been proved to have significant regulatory effect on the immune rejection after hematopoietic stem cell transplantation, its role in solid-organ transplantation remains uncovered. In this study, we investigate whether histone methylation regulation can impact renal allograft rejection in rat models. RESULTS Allogeneic rat renal transplantation model (Wistar to Lewis) was established, and the recipients were administrated with EZH2 inhibitor DZNep after transplantation. Renal allografts and peripheral blood were collected on day 5 after transplantation for histological examination and mechanism investigation. We found that inhibition of EZH2 by DZNep after transplantation significantly ameliorated acute rejection (AR), with decreased histological injury and reduced inflammatory infiltration in renal allografts. Attenuation of AR was due to the prohibited activation of alloreactive T cells, the subsequent impaired production of inflammatory cytokines, and also the elevated apoptosis of alloreactive T cells in both renal allografts and periphery. However, inhibition of EZH2 did not increase the regulatory T cells during the AR. CONCLUSIONS Disruption of EZH2 by DZNep suppressed the immune responses of alloreactive T cells and ameliorated AR of renal allografts. This suggests a therapeutic potential of targeting histone methyltransferases EZH2 in treating allograft rejection after solid organ transplantation.
Collapse
Affiliation(s)
- Long Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Yi Zhang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China.,Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Jina Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China.
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China.
| |
Collapse
|
97
|
Safa K, Chandran S, Wojciechowski D. Pharmacologic targeting of regulatory T cells for solid organ transplantation: current and future prospects. Drugs 2016; 75:1843-52. [PMID: 26493288 DOI: 10.1007/s40265-015-0487-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The last three decades have witnessed significant advances in the development of immunosuppressive medications used in kidney transplantation leading to a remarkable gain in short-term graft function and outcomes. Despite these major breakthroughs, improvements in long-term outcomes lag behind due to a stalemate between drug-related nephrotoxicity and chronic rejection typically due to donor-specific antibodies. Regulatory T cells (Tregs) have been shown to modulate the alloimmune response and can exert suppressive activity preventing allograft rejection in kidney transplantation. Currently available immunosuppressive agents impact Tregs in the alloimmune milieu with some of these interactions being deleterious to the allograft while others may be beneficial. Variable effects are seen with common antibody induction agents such that basiliximab, an IL-2 receptor blocker, decreases Tregs while lymphocyte depleting agents such as antithymocyte globulin increase Tregs. Calcineurin inhibitors, a mainstay of maintenance immunosuppression since the mid-1980s, seem to suppress Tregs while mammalian targets of rapamycin (less commonly used in maintenance regimens) expand Tregs. The purpose of this review is to provide an overview of Treg biology in transplantation, identify in more detail the interactions between commonly used immunosuppressive agents and Tregs in kidney transplantation and lastly describe future directions in the use of Tregs themselves as therapy for tolerance induction.
Collapse
Affiliation(s)
- Kassem Safa
- Division of Nephrology and Transplant Center, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA, USA
| | - Sindhu Chandran
- Division of Nephrology, Department of Medicine, University of California San Francisco Medical center, San Francisco, CA, USA
| | - David Wojciechowski
- Division of Nephrology and Transplant Center, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA, USA.
| |
Collapse
|
98
|
Baroja-Mazo A, Revilla-Nuin B, Parrilla P, Martínez-Alarcón L, Ramírez P, Pons JA. Tolerance in liver transplantation: Biomarkers and clinical relevance. World J Gastroenterol 2016; 22:7676-91. [PMID: 27678350 PMCID: PMC5016367 DOI: 10.3748/wjg.v22.i34.7676] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/04/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
Transplantation is the optimal treatment for end-stage organ failure, and modern immunosuppression has allowed important progress in short-term outcomes. However, immunosuppression poorly influences chronic rejection and elicits chronic toxicity in current clinical practice. Thus, a major goal in transplantation is to understand and induce tolerance. It is well established that human regulatory T cells expressing the transcription factor FoxP3 play important roles in the maintenance of immunological self-tolerance and immune homeostasis. The major regulatory T cell subsets and mechanisms of expansion that are critical for induction and long-term maintenance of graft tolerance and survival are being actively investigated. Likewise, other immune cells, such as dendritic cells, monocyte/macrophages or natural killer cells, have been described as part of the process known as "operational tolerance". However, translation of these results towards clinical practice needs solid tools to identify accurately and reliably patients who are going to be tolerant. In this way, a plethora of genetic and cellular biomarkers is raising and being validated worldwide in large multi-center clinical trials. Few of the studies performed so far have provided a detailed analysis of the impact of immunosuppression withdrawal on pre-existing complications derived from the long-term administration of immunosuppressive drugs and the side effects associated with them. The future of liver transplantation is aimed to develop new therapies which increase the actual low tolerant vs non-tolerant recipients ratio.
Collapse
|
99
|
Hu M, Wang YM, Wang Y, Zhang GY, Zheng G, Yi S, O'Connell PJ, Harris DCH, Alexander SI. Regulatory T cells in kidney disease and transplantation. Kidney Int 2016; 90:502-514. [PMID: 27263492 DOI: 10.1016/j.kint.2016.03.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/06/2016] [Accepted: 03/17/2016] [Indexed: 01/03/2023]
Abstract
Regulatory T cells (Tregs) have been shown to be important in maintaining immune homeostasis and preventing autoimmune disease, including autoimmune kidney disease. It is also likely that they play a role in limiting kidney transplant rejection and potentially in promoting transplant tolerance. Although other subsets of Tregs exist, the most potent and well-defined Tregs are the Foxp3 expressing CD4(+) Tregs derived from the thymus or generated peripherally. These CD4(+)Foxp3(+) Tregs limit autoimmune renal disease in animal models, especially chronic kidney disease, and kidney transplantation. Furthermore, other subsets of Tregs, including CD8 Tregs, may play a role in immunosuppression in kidney disease. The development and protective mechanisms of Tregs in kidney disease and kidney transplantation involve multiple mechanisms of suppression. Here we review the development and function of CD4(+)Foxp3(+) Tregs. We discuss the specific application of Tregs as a therapeutic strategy to prevent kidney disease and to limit kidney transplant rejection and detail clinical trials in this area of transplantation.
Collapse
Affiliation(s)
- Min Hu
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia; Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Yiping Wang
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Geoff Y Zhang
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Shounan Yi
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Philip J O'Connell
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - David C H Harris
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia.
| |
Collapse
|
100
|
Chen D, Li Y, Wang X, Li K, Jing Y, He J, Qiang Z, Tong J, Sun K, Ding W, Kang Y, Li G. Generation of regulatory dendritic cells after treatment with paeoniflorin. Immunol Res 2016; 64:988-1000. [PMID: 26721806 DOI: 10.1007/s12026-015-8773-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Regulatory dendritic cells are a potential therapeutic tool for assessing a variety of immune overreaction diseases. Paeoniflorin, a bioactive glucoside extracted from the Chinese herb white paeony root, has been shown to be effective at inhibiting the maturation and immunostimulatory function of murine bone marrow-derived dendritic cells. However, whether paeoniflorin can program conventional dendritic cells toward regulatory dendritic cells and the underlying mechanism remain unknown. Here, our study demonstrates that paeoniflorin can induce the production of regulatory dendritic cells from human peripheral blood monocyte-derived immature dendritic cells in the absence or presence of lipopolysaccharide (LPS) but not from mature dendritic cells, thereby demonstrating the potential of paeoniflorin as a specific immunosuppressive drug with fewer complications and side effects. These regulatory dendritic cells treated with paeoniflorin exhibited high CD11b/c and low CD80, CD86 and CD40 expression levels as well as enhanced abilities to capture antigen and promote the proliferation of CD4(+)CD25(+) T cells and reduced abilities to migrate and promote the proliferation of CD4(+) T cells, which is associated with the upregulation of endogenous transforming growth factor (TGF)-β-mediated indoleamine 2,3-dioxygenase (IDO) expression. Collectively, paeoniflorin could program immature dendritic cells (imDCs) and imDCs stimulated with LPS toward a regulatory DC fate by upregulating the endogenous TGF-β-mediated IDO expression level, thereby demonstrating its potential as a specific immunosuppressive drug.
Collapse
Affiliation(s)
- Dan Chen
- Department of Pharmacology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yingxi Li
- Department of Biology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Xiaodong Wang
- Department of Pharmacology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Keqiu Li
- Department of Biology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yaqing Jing
- Department of Biology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Jinghua He
- Department of Pharmacology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Zhaoyan Qiang
- Department of Pharmacology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Jingzhi Tong
- Department of Pharmacology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Ke Sun
- Department of Pharmacology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Wen Ding
- Department of Pharmacology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yi Kang
- Department of Pharmacology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China.
| | - Guang Li
- Department of Biology, School of Basic Medical Sciences, Basic Medical College, Tianjin Medical University, Tianjin, 300070, People's Republic of China.
| |
Collapse
|