1
|
Knoedler L, Dean J, Diatta F, Thompson N, Knoedler S, Rhys R, Sherwani K, Ettl T, Mayer S, Falkner F, Kilian K, Panayi AC, Iske J, Safi AF, Tullius SG, Haykal S, Pomahac B, Kauke-Navarro M. Immune modulation in transplant medicine: a comprehensive review of cell therapy applications and future directions. Front Immunol 2024; 15:1372862. [PMID: 38650942 PMCID: PMC11033354 DOI: 10.3389/fimmu.2024.1372862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024] Open
Abstract
Balancing the immune response after solid organ transplantation (SOT) and vascularized composite allotransplantation (VCA) remains an ongoing clinical challenge. While immunosuppressants can effectively reduce acute rejection rates following transplant surgery, some patients still experience recurrent acute rejection episodes, which in turn may progress to chronic rejection. Furthermore, these immunosuppressive regimens are associated with an increased risk of malignancies and metabolic disorders. Despite significant advancements in the field, these IS related side effects persist as clinical hurdles, emphasizing the need for innovative therapeutic strategies to improve transplant survival and longevity. Cellular therapy, a novel therapeutic approach, has emerged as a potential pathway to promote immune tolerance while minimizing systemic side-effects of standard IS regiments. Various cell types, including chimeric antigen receptor T cells (CAR-T), mesenchymal stromal cells (MSCs), regulatory myeloid cells (RMCs) and regulatory T cells (Tregs), offer unique immunomodulatory properties that may help achieve improved outcomes in transplant patients. This review aims to elucidate the role of cellular therapies, particularly MSCs, T cells, Tregs, RMCs, macrophages, and dendritic cells in SOT and VCA. We explore the immunological features of each cell type, their capacity for immune regulation, and the prospective advantages and obstacles linked to their application in transplant patients. An in-depth outline of the current state of the technology may help SOT and VCA providers refine their perioperative treatment strategies while laying the foundation for further trials that investigate cellular therapeutics in transplantation surgery.
Collapse
Affiliation(s)
- Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fortunay Diatta
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Noelle Thompson
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Samuel Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Richmond Rhys
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Khalil Sherwani
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Tobias Ettl
- Department of Dental, Oral and Maxillofacial Surgery, Regensburg, Germany
| | - Simon Mayer
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Florian Falkner
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Katja Kilian
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Adriana C. Panayi
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Jasper Iske
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ali-Farid Safi
- Faculty of Medicine, University of Bern, Bern, Switzerland
- Craniologicum, Center for Cranio-Maxillo-Facial Surgery, Bern, Switzerland
| | - Stefan G. Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Siba Haykal
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bohdan Pomahac
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Martin Kauke-Navarro
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
2
|
Mittal SK, Cho W, Elbasiony E, Guan Y, Foulsham W, Chauhan SK. Mesenchymal stem cells augment regulatory T cell function via CD80-mediated interactions and promote allograft survival. Am J Transplant 2022; 22:1564-1577. [PMID: 35170213 PMCID: PMC11261724 DOI: 10.1111/ajt.17001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023]
Abstract
Mesenchymal stem cells (MSCs) and regulatory T cells (Tregs) both have been shown to modulate the alloimmune response and promote transplant survival. Mounting evidence suggests that MSCs augment Treg function, but the mechanisms underlying this phenomenon have not been fully deciphered. Here, we identified that MSCs express substantial levels of CD80 and evaluated its immunoregulatory function using in vivo and in vitro experiments. Our in vitro culture assays demonstrated that MSCs induce expression of FoxP3 in Tregs in a contact-dependent manner, and the blockade of CD80 abrogates this FoxP3 induction and Treg-mediated suppression of T cell proliferation. Moreover, supplementation of soluble CD80 significantly upregulated FoxP3 expression. Using a well-characterized murine model of corneal transplantation, we show that silencing CD80 in MSCs diminishes the capacity of MSCs to promote selective graft infiltration of Tregs, promote FoxP3 expression and upregulate suppressive function of Tregs. Consequently, MSCs, following CD80 knockdown, failed to promote corneal allograft survival.
Collapse
Affiliation(s)
- Sharad K Mittal
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - WonKyung Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Yilin Guan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - William Foulsham
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Hefazi M, Bolivar-Wagers S, Blazar BR. Regulatory T Cell Therapy of Graft-versus-Host Disease: Advances and Challenges. Int J Mol Sci 2021; 22:9676. [PMID: 34575843 PMCID: PMC8469916 DOI: 10.3390/ijms22189676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/03/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
Graft-versus-host disease (GVHD) is the leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Immunomodulation using regulatory T cells (Tregs) offers an exciting option to prevent and/or treat GVHD as these cells naturally function to maintain immune homeostasis, can induce tolerance following HSCT, and have a tissue reparative function. Studies to date have established a clinical safety profile for polyclonal Tregs. Functional enhancement through genetic engineering offers the possibility of improved potency, specificity, and persistence. In this review, we provide the most up to date preclinical and clinical data on Treg cell therapy with a particular focus on GVHD. We discuss the different Treg subtypes and highlight the pharmacological and genetic approaches under investigation to enhance the application of Tregs in allo-HSCT. Lastly, we discuss the remaining challenges for optimal clinical translation and provide insights as to future directions of the field.
Collapse
Affiliation(s)
- Mehrdad Hefazi
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Sara Bolivar-Wagers
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA;
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA;
| |
Collapse
|
4
|
Abstract
Involvement of T lymphocytes in kidney transplantation is a well-developed topic; however, most of the scientific interest focused on the different type of CD4+ lymphocyte subpopulations. Few authors, instead, investigated the role of CD8+ T cells in renal transplantation and how deleterious they can be to long-term allograft survival. Recently, there has been a renewed interest in the CD8+ T cells involvement in the transplantation field with the aim to investigate the immunological mechanisms underlying the infiltration of CD8+ T cells and their biological functions in human kidney allografts. The purpose of the present review is to highlight the role of allo-reactive cytotoxic T lymphocytes (CTLs) CD8+ subset in allograft kidney recipients and their related clinical complications.
Collapse
|
5
|
Moorman CD, Sohn SJ, Phee H. Emerging Therapeutics for Immune Tolerance: Tolerogenic Vaccines, T cell Therapy, and IL-2 Therapy. Front Immunol 2021; 12:657768. [PMID: 33854514 PMCID: PMC8039385 DOI: 10.3389/fimmu.2021.657768] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases affect roughly 5-10% of the total population, with women affected more than men. The standard treatment for autoimmune or autoinflammatory diseases had long been immunosuppressive agents until the advent of immunomodulatory biologic drugs, which aimed at blocking inflammatory mediators, including proinflammatory cytokines. At the frontier of these biologic drugs are TNF-α blockers. These therapies inhibit the proinflammatory action of TNF-α in common autoimmune diseases such as rheumatoid arthritis, psoriasis, ulcerative colitis, and Crohn's disease. TNF-α blockade quickly became the "standard of care" for these autoimmune diseases due to their effectiveness in controlling disease and decreasing patient's adverse risk profiles compared to broad-spectrum immunosuppressive agents. However, anti-TNF-α therapies have limitations, including known adverse safety risk, loss of therapeutic efficacy due to drug resistance, and lack of efficacy in numerous autoimmune diseases, including multiple sclerosis. The next wave of truly transformative therapeutics should aspire to provide a cure by selectively suppressing pathogenic autoantigen-specific immune responses while leaving the rest of the immune system intact to control infectious diseases and malignancies. In this review, we will focus on three main areas of active research in immune tolerance. First, tolerogenic vaccines aiming at robust, lasting autoantigen-specific immune tolerance. Second, T cell therapies using Tregs (either polyclonal, antigen-specific, or genetically engineered to express chimeric antigen receptors) to establish active dominant immune tolerance or T cells (engineered to express chimeric antigen receptors) to delete pathogenic immune cells. Third, IL-2 therapies aiming at expanding immunosuppressive regulatory T cells in vivo.
Collapse
Affiliation(s)
| | | | - Hyewon Phee
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| |
Collapse
|
6
|
Bayati F, Mohammadi M, Valadi M, Jamshidi S, Foma AM, Sharif-Paghaleh E. The Therapeutic Potential of Regulatory T Cells: Challenges and Opportunities. Front Immunol 2021; 11:585819. [PMID: 33519807 PMCID: PMC7844143 DOI: 10.3389/fimmu.2020.585819] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Tregs) are an immunosuppressive subgroup of CD4+ T cells which are identified by the expression of forkhead box protein P3 (Foxp3). The modulation capacity of these immune cells holds an important role in both transplantation and the development of autoimmune diseases. These cells are the main mediators of self-tolerance and are essential for avoiding excessive immune reactions. Tregs play a key role in the induction of peripheral tolerance that can prevent autoimmunity, by protecting self-reactive lymphocytes from the immune reaction. In contrast to autoimmune responses, tumor cells exploit Tregs in order to prevent immune cell recognition and anti-tumor immune response during the carcinogenesis process. Recently, numerous studies have focused on unraveling the biological functions and principles of Tregs and their primary suppressive mechanisms. Due to the promising and outstanding results, Tregs have been widely investigated as an alternative tool in preventing graft rejection and treating autoimmune diseases. On the other hand, targeting Tregs for the purpose of improving cancer immunotherapy is being intensively evaluated as a desirable and effective method. The purpose of this review is to point out the characteristic function and therapeutic potential of Tregs in regulatory immune mechanisms in transplantation tolerance, autoimmune diseases, cancer therapy, and also to discuss that how the manipulation of these mechanisms may increase the therapeutic options.
Collapse
Affiliation(s)
- Fatemeh Bayati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research & Development Department, Aryogen Pharmed, Karaj, Iran
| | - Mahsa Mohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Maryam Valadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Jamshidi
- Research & Development Department, Aryogen Pharmed, Karaj, Iran
| | - Arron Munggela Foma
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Sharif-Paghaleh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
7
|
López-Abente J, Martínez-Bonet M, Bernaldo-de-Quirós E, Camino M, Gil N, Panadero E, Gil-Jaurena JM, Clemente M, Urschel S, West L, Pion M, Correa-Rocha R. Basiliximab impairs regulatory T cell (TREG) function and could affect the short-term graft acceptance in children with heart transplantation. Sci Rep 2021; 11:827. [PMID: 33436905 PMCID: PMC7803770 DOI: 10.1038/s41598-020-80567-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/21/2020] [Indexed: 12/26/2022] Open
Abstract
CD25, the alpha chain of the IL-2 receptor, is expressed on activated effector T cells that mediate immune graft damage. Induction immunosuppression is commonly used in solid organ transplantation and can include antibodies blocking CD25. However, regulatory T cells (Tregs) also rely on CD25 for their proliferation, survival, and regulatory function. Therefore, CD25-blockade may compromise Treg protective role against rejection. We analysed in vitro the effect of basiliximab (BXM) on the viability, phenotype, proliferation and cytokine production of Treg cells. We also evaluated in vivo the effect of BXM on Treg in thymectomized heart transplant children receiving BXM in comparison to patients not receiving induction therapy. Our results show that BXM reduces Treg counts and function in vitro by affecting their proliferation, Foxp3 expression, and IL-10 secretion capacity. In pediatric heart-transplant patients, we observed decreased Treg counts and a diminished Treg/Teff ratio in BXM-treated patients up to 6-month after treatment, recovering baseline values at the end of the 12-month follow up period. These results reveal that the use of BXM could produce detrimental effects on Tregs, and support the evidence suggesting that BXM induction could impair the protective role of Tregs in the period of highest incidence of acute graft rejection.
Collapse
Affiliation(s)
- Jacobo López-Abente
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain
| | - Marta Martínez-Bonet
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain
| | - Esther Bernaldo-de-Quirós
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain
| | - Manuela Camino
- Pediatric-Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Nuria Gil
- Pediatric-Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Esther Panadero
- Pediatric-Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Juan Miguel Gil-Jaurena
- Pediatric Cardiac Surgery Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Maribel Clemente
- Cell Culture Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Simon Urschel
- Pediatric Cardiac Transplantation, University of Alberta/Stollery Children's Hospital, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada.,Canadian National Transplant Research Program Investigator, CNTRP, Edmonton, AB, Canada
| | - Lori West
- Pediatric Cardiac Transplantation, University of Alberta/Stollery Children's Hospital, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada.,Canadian National Transplant Research Program Investigator, CNTRP, Edmonton, AB, Canada
| | - Marjorie Pion
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain. .,Canadian National Transplant Research Program Investigator, CNTRP, Edmonton, AB, Canada.
| |
Collapse
|
8
|
Chen AC, Cai X, Li C, Khoryati L, Gavin MA, Miao CH. A Treg-Selective IL-2 Mutein Prevents the Formation of Factor VIII Inhibitors in Hemophilia Mice Treated With Factor VIII Gene Therapy. Front Immunol 2020; 11:638. [PMID: 32411127 PMCID: PMC7198749 DOI: 10.3389/fimmu.2020.00638] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
Hemophilia A is a genetic disorder that results in the deficiency of functional factor VIII protein, which plays a key role in blood coagulation. Currently, the majority of hemophilia A patients are treated with repeated infusions of factor VIII protein. Approximately 30% of severe hemophilia A patients develop neutralizing antibodies to factor VIII (known as factor VIII inhibitors) due to treatment, rendering factor VIII protein infusions ineffective. Previously, mice receiving murine IL-2 complexed with α-murine IL-2 mAbs (JES6-1A12) showed a lack of factor VIII inhibitor formation after factor VIII treatment, which was associated with the proliferation and the activation of factor VIII-specific regulatory T cells (Tregs). In this paper, we evaluated if an Fc-fused mutated protein analog of mouse IL-2, named Fc.Mut24, engineered to selectively promote the expansion of Tregs in vivo can modulate factor VIII-specific immune responses. The mice received one intraperitoneal injection of Fc.Mut24. When the regulatory T cell population reached its highest frequency and peak activation, the mice received a hydrodynamic injection of factor VIII plasmid (day 4) followed by a second Fc.Mut24 dose (day 7). Peripheral blood was collected weekly. Flow cytometry was used to characterize the peripheral blood cell populations, while ELISA and Bethesda assays were used to assess the inhibitor concentrations and the functional titers in plasma. The activated partial thromboplastin time assay was used to assess the functional activities of factor VIII in blood. The mice receiving Fc.Mut24 showed a dramatic and transient increase in the population of activated Tregs after Fc.Mut24 injection. Factor VIII gene therapy via hydrodynamic injection resulted in high anti-factor VIII inhibitor concentrations in control PBS-injected mice, whereas the mice treated with Fc.Mut24 produced no inhibitors. Most significantly, there were no inhibitors generated after a second hydrodynamic injection of factor VIII plasmid administered at 19 weeks after the first injection in Fc.Mut24-treated mice. The mice receiving Fc.Mut24 maintained high levels of factor VIII activity throughout the experiment, while the control mice had the factor VIII activity dropped to undetectable levels a few weeks after the first factor VIII plasmid injection. Our data show that human therapies analogous to Fc.Mut24 could potentially provide a method to prevent inhibitor formation and induce long-term immune tolerance to factor VIII in hemophilia patients.
Collapse
Affiliation(s)
- Alex C. Chen
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Xiaohe Cai
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Chong Li
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Liliane Khoryati
- Translational Research Program, Benaroya Research Institute, Seattle, WA, United States
| | - Marc A. Gavin
- Translational Research Program, Benaroya Research Institute, Seattle, WA, United States
| | - Carol H. Miao
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, United States
| |
Collapse
|
9
|
Mandatori S, Pacella I, Marzolla V, Mammi C, Starace D, Padula F, Vitiello L, Armani A, Savoia C, Taurino M, De Zio D, Giampietri C, Piconese S, Cecconi F, Caprio M, Filippini A. Altered Tregs Differentiation and Impaired Autophagy Correlate to Atherosclerotic Disease. Front Immunol 2020; 11:350. [PMID: 32231663 PMCID: PMC7082762 DOI: 10.3389/fimmu.2020.00350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/13/2020] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is a progressive vascular disease representing the primary cause of morbidity and mortality in developed countries. Formerly, atherosclerosis was considered as a mere passive accumulation of lipids in blood vessels. However, it is now clear that atherosclerosis is a complex and multifactorial disease, in which the involvement of immune cells and inflammation play a key role. A variety of studies have shown that autophagy-a cellular catalytic mechanism able to remove injured cytoplasmic components in response to cellular stress-may be proatherogenic. So far, in this context, its role has been investigated in smooth muscle cells, macrophages, and endothelial cells, while the function of this catabolic protective process in lymphocyte functionality has been overlooked. The few studies carried out so far, however, suggested that autophagy modulation in lymphocyte subsets may be functionally related to plaque formation and development. Therefore, in this research, we aimed at better clarifying the role of lymphocyte subsets, mainly regulatory T cells (Tregs), in human atherosclerotic plaques and in animal models of atherosclerosis investigating the contribution of autophagy on immune cell homeostasis. Here, we investigate basal autophagy in a mouse model of atherosclerosis, apolipoprotein E (ApoE)-knockout (KO) mice, and we analyze the role of autophagy in driving Tregs polarization. We observed defective maturation of Tregs from ApoE-KO mice in response to tumor growth factor-β (TGFβ). TGFβ is a well-known autophagy inducer, and Tregs maturation defects in ApoE-KO mice seem to be related to autophagy impairment. In this work, we propose that autophagy underlies Tregs maturation, advocating that the study of this process in atherosclerosis may open new therapeutic strategies.
Collapse
Affiliation(s)
- Sara Mandatori
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| | - Ilenia Pacella
- Laboratory of Cellular and Molecular Immunology, Department of Internal Clinical Sciences, Anaesthesiology and Cardiovascular Sciences, Sapienza Università di Roma, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Caterina Mammi
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Donatella Starace
- UOC, Clinical Pathology, San Giovanni Addolorata Hospital, Rome, Italy
| | - Fabrizio Padula
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| | - Laura Vitiello
- Flow Cytometry Unit, IRCCS San Raffaele Pisana, Rome, Italy
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Carmine Savoia
- Cardiology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Maurizio Taurino
- Unit of Vascular Surgery, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Daniela De Zio
- Cell Stress and Survival Unit, Center of Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Claudia Giampietri
- Unit of Human Anatomy, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| | - Silvia Piconese
- Laboratory of Cellular and Molecular Immunology, Department of Internal Clinical Sciences, Anaesthesiology and Cardiovascular Sciences, Sapienza Università di Roma, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Francesco Cecconi
- Cell Stress and Survival Unit, Center of Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Paediatric Haematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| | - Antonio Filippini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Lim EL, Okkenhaug K. Phosphoinositide 3-kinase δ is a regulatory T-cell target in cancer immunotherapy. Immunology 2019; 157:210-218. [PMID: 31107985 PMCID: PMC6587315 DOI: 10.1111/imm.13082] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/02/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022] Open
Abstract
Tumour infiltration by regulatory T (Treg) cells contributes to suppression of the anti-tumour immune response, which limits the efficacy of immune-mediated cancer therapies. The phosphoinositide 3-kinase (PI3K) pathway has key roles in mediating the function of many immune cell subsets, including Treg cells. Treg function is context-dependent and depends on input from different cell surface receptors, many of which can activate the PI3K pathway. In this review, we explore how PI3Kδ contributes to signalling through several major immune cell receptors, including the T-cell receptor and co-stimulatory receptors such as CD28 and ICOS, but is antagonized by the immune checkpoint receptors CTLA-4 and PD-1. Understanding how PI3Kδ inhibition affects Treg signalling events will help to inform how best to use PI3Kδ inhibitors in clinical cancer treatment.
Collapse
Affiliation(s)
- Ee Lyn Lim
- Laboratory of Experimental ImmunologyImmunology Frontier Research CentreOsaka UniversitySuitaJapan
| | - Klaus Okkenhaug
- Division of ImmunologyDepartment of PathologyUniversity of CambridgeCambridgeUK
| |
Collapse
|
11
|
Thangavelu G, Blazar BR. Achievement of Tolerance Induction to Prevent Acute Graft-vs.-Host Disease. Front Immunol 2019; 10:309. [PMID: 30906290 PMCID: PMC6419712 DOI: 10.3389/fimmu.2019.00309] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/06/2019] [Indexed: 01/04/2023] Open
Abstract
Acute graft-vs.-host disease (GVHD) limits the efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT), a main therapy to treat various hematological disorders. Despite rapid progress in understanding GVHD pathogenesis, broad immunosuppressive agents are most often used to prevent and remain the first line of therapy to treat GVHD. Strategies enhancing immune tolerance in allo-HSCT would permit reductions in immunosuppressant use and their associated undesirable side effects. In this review, we discuss the mechanisms responsible for GVHD and advancement in strategies to achieve immune balance and tolerance thereby avoiding GVHD and its complications.
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
12
|
Tahvildari M, Inomata T, Amouzegar A, Dana R. Regulatory T cell modulation of cytokine and cellular networks in corneal graft rejection. CURRENT OPHTHALMOLOGY REPORTS 2018; 6:266-274. [PMID: 31807370 PMCID: PMC6894425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
PURPOSE OF REVIEW Corneal allografts placed in vascularized or inflamed host beds are at increased risk of graft rejection due to the preponderance of activated immune cells in the host bed. Regulatory T cells (Tregs) are master regulators of the adaptive immune response and play a key role in the induction of immune tolerance. The aim of this review is to discuss mechanisms through which Tregs mediate tolerance in corneal transplantation and the novel therapeutic approaches that target Tregs to promote transplant survival. RECENT FINDINGS The inflammatory environment of high-risk allografts not only promotes activation of effector T cells and their infiltration to graft site, but also impairs Treg immunomodulatory function. Recent studies have shown that expansion of Tregs and enhancing their modulatory function significantly improve graft survival. SUMMARY As our understanding of the cellular and molecular pathways in corneal transplantation has deepened, novel therapeutic strategies have been developed to improve allograft survival. In this review, we discuss therapeutic approaches that focus on Tregs to promote corneal allograft survival.
Collapse
Affiliation(s)
- Maryam Tahvildari
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA
- Kresge Eye Institute, Department of ophthalmology, Wayne State University, Detroit, MI
| | - Takenori Inomata
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA
- Juntendo University Faculty of Medicine, Department of Ophthalmology, Tokyo, Japan
- Juntendo University Faculty of Medicine, Department of Strategic Operative Room, Management and Improvement, Tokyo, Japan
| | - Afsaneh Amouzegar
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA
| |
Collapse
|
13
|
Tahvildari M, Inomata T, Amouzegar A, Dana R. Regulatory T Cell Modulation of Cytokine and Cellular Networks in Corneal Graft Rejection. CURRENT OPHTHALMOLOGY REPORTS 2018. [DOI: 10.1007/s40135-018-0191-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Delayed allogeneic skin graft rejection in CD26-deficient mice. Cell Mol Immunol 2018; 16:557-567. [PMID: 29572550 PMCID: PMC6804736 DOI: 10.1038/s41423-018-0009-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 12/21/2022] Open
Abstract
Organ transplantation is an effective therapeutic tool for treating many terminal diseases. However, one of the biggest challenges of transplantation is determining how to achieve the long-term survival of the allogeneic or xenogeneic transplant by, for example, preventing transplant rejection. In the current study, CD26 gene-knockout mice were used to investigate the potential role of CD26/dipeptidyl peptidase-4 (DPPIV) in allogeneic skin graft rejection by tail-skin transplantation. Compared with wild-type (CD26+/+) counterparts, CD26-/- mice showed reduced necrosis of grafts and delayed graft rejection after skin transplantation. Concentrations of serum IgG, including its subclasses IgG1 and IgG2a, were significantly reduced in CD26-/- mice during graft rejection. Moreover, after allogeneic skin transplantation, the secretion levels of the cytokines IFN-γ, IL-2, IL-6, IL-4, and IL-13 were significantly reduced, whereas the level of the cytokine IL-10 was increased in the serum of CD26-/- mice compared with that in the serum of CD26+/+ mice. Additionally, the concentration of IL-17 in serum and the percentage of cells secreting IL-17 in mouse peripheral blood lymphocytes (MPBLs) were both significantly lower, while the percentage of regulatory T cells (Tregs) was significantly higher in MPBLs of CD26-/- mice than in those of CD26+/+ mice. Furthermore, a lower percentage of CD8+ T cells in MPBLs and fewer infiltrated macrophages and T cells in graft tissues of CD26-/- mice were detected during graft rejection. These results indicate that CD26 is involved in allogeneic skin graft rejection and provides another hint that CD26 deficiency leads to less rejection due to lower activation and proliferation of host immune cells.
Collapse
|
15
|
Oyarce K, Campos-Mora M, Gajardo-Carrasco T, Pino-Lagos K. Vitamin C Fosters the In Vivo Differentiation of Peripheral CD4 + Foxp3 - T Cells into CD4 + Foxp3 + Regulatory T Cells but Impairs Their Ability to Prolong Skin Allograft Survival. Front Immunol 2018; 9:112. [PMID: 29479348 PMCID: PMC5811461 DOI: 10.3389/fimmu.2018.00112] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/15/2018] [Indexed: 12/16/2022] Open
Abstract
Regulatory T cells (Tregs) are critical players of immunological tolerance due to their ability to suppress effector T cell function thereby preventing transplant rejection and autoimmune diseases. During allograft transplantation, increases of both Treg expansion and generation, as well as their stable function, are needed to ensure allograft acceptance; thus, efforts have been made to discover new molecules that enhance Treg-mediated tolerance and to uncover their mechanisms. Recently, vitamin C (VitC), known to regulate T cell maturation and dendritic cell-mediated T cell polarization, has gained attention as a relevant epigenetic remodeler able to enhance and stabilize the expression of the Treg master regulator gene Foxp3, positively affecting the generation of induced Tregs (iTregs). In this study, we measured VitC transporter (SVCT2) expression in different immune cell populations, finding Tregs as one of the cell subset with the highest levels of SVCT2 expression. Unexpectedly, we found that VitC treatment reduces the ability of natural Tregs to suppress effector T cell proliferation in vitro, while having an enhancer effect on TGFβ-induced Foxp3+ Tregs. On the other hand, VitC increases iTregs generation in vitro and in vivo, however, no allograft tolerance was achieved in animals orally treated with VitC. Lastly, Tregs isolated from the draining lymph nodes of VitC-treated and transplanted mice also showed impaired suppression capacity ex vivo. Our results indicate that VitC promotes the generation and expansion of Tregs, without exhibiting CD4+ T cell-mediated allograft tolerance. These observations highlight the relevance of the nutritional status of patients when immune regulation is needed.
Collapse
Affiliation(s)
- Karina Oyarce
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Mauricio Campos-Mora
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Tania Gajardo-Carrasco
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Karina Pino-Lagos
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
16
|
Zhang W, Li J, Qi G, Tu G, Yang C, Xu M. Myeloid-derived suppressor cells in transplantation: the dawn of cell therapy. J Transl Med 2018; 16:19. [PMID: 29378596 PMCID: PMC5789705 DOI: 10.1186/s12967-018-1395-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 01/22/2018] [Indexed: 02/07/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a series of innate cells that play a significant role in inhibiting T cell-related responses. This heterogeneous population of immature cells is involved in tumor immunity. Recently, the function and importance of MDSCs in transplantation have garnered the attention of scientists and have become an important focus of transplantation immunology research because MDSCs play a key role in establishing immune tolerance in transplantation. In this review, we summarize recent studies of MDSCs in different types of transplantation. We also focus on the influence of immunosuppressive drugs on MDSCs as well as future obstacles and research directions in this field.
Collapse
Affiliation(s)
- Weitao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Jiawei Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Guisheng Qi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Guowei Tu
- Department of Intensive Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| |
Collapse
|
17
|
Kodati S, Chauhan SK, Chen Y, Emami-Naeini P, Omoto M, Dohlman TH, Stevenson W, Amouzegar A, Tummala G, Saban DR, Dana R. Interleukin-6 neutralization prolongs corneal allograft survival. CURRENT TRENDS IN IMMUNOLOGY 2018; 19:105-113. [PMID: 30906117 PMCID: PMC6428205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The purpose of this study is to determine the effect of systemic blockade of Interleukin-6 (IL-6) on allosensitization, regulatory T cell frequencies and suppressive phenotype, and allograft survival rates in a mouse model of corneal transplantation. Allogeneic corneal transplantation was performed using C57BL/6 mice as donors and BALB/c mice as recipients. Graft recipients were injected daily with either anti-IL-6 antibody or an isotype control antibody (1.25 mg/ml) for the first 7 days and on alternate days thereafter until week 8 after transplantation. Allograft survival was evaluated for 8 weeks using Kaplan-Meier survival analysis. Draining lymph nodes (DLN) were harvested at week 3 after transplantation, and proliferation of isolated recipient T cells through direct and indirect pathways was determined using mixed lymphocyte reaction assay. Frequencies of CD4+CD25+Foxp3+ regulatory T cells, their expression of Foxp3, and frequencies of IFNy+CD4+ Th1 cells were determined in DLN using flow cytometry. Finally, CD4+ T cells were cultured with bone marrow-derived dendritic cells from either C57BL/6 or BALB/c mice in the presence of IL-6-blocking antibody to determine Treg induction through direct and indirect pathways, respectively. Treatment with anti-IL-6 antibody suppressed both the direct and indirect pathways of allosensitization in graft recipients and significantly improved allograft survival rates. Furthermore, in vivo blockade of IL-6 enhanced Foxp3 expression by Tregs in graft recipients undergoing rejection, but did not exert a significant effect on Treg frequencies. Finally, IL-6 neutralization in vitro enhanced the differentiation of Tregs from CD4+ T cells through both direct and indirect pathways. Our results demonstrate that systemic administration of IL-6-blocking antibody to corneal allograft recipients suppresses direct and indirect routes of allosensitization, is associated with increased expression of Foxp3 by Tregs, and improves allograft survival rates.
Collapse
|
18
|
Sula Karreci E, Eskandari SK, Dotiwala F, Routray SK, Kurdi AT, Assaker JP, Luckyanchykov P, Mihali AB, Maarouf O, Borges TJ, Alkhudhayri A, Patel KR, Radwan A, Ghobrial I, McGrath M, Chandraker A, Riella LV, Elyaman W, Abdi R, Lieberman J, Azzi J. Human regulatory T cells undergo self-inflicted damage via granzyme pathways upon activation. JCI Insight 2017; 2:91599. [PMID: 29093262 DOI: 10.1172/jci.insight.91599] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 09/25/2017] [Indexed: 12/13/2022] Open
Abstract
Tregs hold great promise as a cellular therapy for multiple immunologically mediated diseases, given their ability to control immune responses. The success of such strategies depends on the expansion of healthy, suppressive Tregs ex vivo and in vivo following the transfer. In clinical studies, levels of transferred Tregs decline sharply in the blood within a few days of the transfer. Tregs have a high rate of apoptosis. Here, we describe a new mechanism of Treg self-inflicted damage. We show that granzymes A and -B (GrA and GrB), which are highly upregulated in human Tregs upon stimulation, leak out of cytotoxic granules to induce cleavage of cytoplasmic and nuclear substrates, precipitating apoptosis in target cells. GrA and GrB substrates were protected from cleavage by inhibiting granzyme activity in vitro. Additionally, we show - by using cytometry by time of flight (CYTOF) - an increase in GrB-expressing Tregs in the peripheral blood and renal allografts of transplant recipients undergoing rejection. These GrB-expressing Tregs showed an activated phenotype but were significantly more apoptotic than non-GrB expressing Tregs. This potentially novel finding improves our understanding of Treg survival and suggests that manipulating Gr expression or activity might be useful for designing more effective Treg therapies.
Collapse
Affiliation(s)
- Esilida Sula Karreci
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Siawosh K Eskandari
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Farokh Dotiwala
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
| | - Sujit K Routray
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Ahmed T Kurdi
- Department of Medical Oncology, Dana-Farber Cancer Institute, and
| | - Jean Pierre Assaker
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Pavlo Luckyanchykov
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Albana B Mihali
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Omar Maarouf
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Thiago J Borges
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Abdullah Alkhudhayri
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Kruti R Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Massachusetts, USA
| | - Amr Radwan
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Irene Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, and
| | - Martina McGrath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Anil Chandraker
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Leonardo V Riella
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Wassim Elyaman
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Massachusetts, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
| | - Jamil Azzi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| |
Collapse
|
19
|
Wu WP, Tsai YG, Lin TY, Wu MJ, Lin CY. The attenuation of renal fibrosis by histone deacetylase inhibitors is associated with the plasticity of FOXP3 +IL-17 + T cells. BMC Nephrol 2017; 18:225. [PMID: 28693431 PMCID: PMC5504832 DOI: 10.1186/s12882-017-0630-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/21/2017] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The histone deacetylase (HDAC) inhibitor, which has potential effects on epigenetic modifications, had been reported to attenuate renal fibrosis. CD4+ forkhead box P3 (FOXP3)+ T regulatory (Treg) cells may be converted to inflammation-associated T helper 17 cells (Th17) with tissue fibrosis properties. The association between FOXP3+IL-17+ T cells and the attenuation of renal fibrosis by the HDAC inhibitor is not clear. METHODS This study evaluated the roles of the HDAC inhibitor, Treg cells and their differentiation into Th17 cells, which aggravate chronic inflammation and renal fibrosis in a unilateral ureteral obstruction (UUO) mouse model. The study groups included control and UUO mice that were monitored for 7, 14 or 21 days. RESULTS Juxtaglomerular (JG) hyperplasia, angiotensin II type 1 receptor (AT1R) expression and lymphocyte infiltration were observed in renal tissues after UUO but were decreased after trichostatin A (TSA) treatment, a HDAC inhibitor. The number of CD4+FOXP3+ T cells increased progressively, along with the number of FOXP3+interleukin (IL)-17+ T cells, after 14 days, and their numbers then progressively decreased with increasing CD4+IL-17+ T cell numbers, as demonstrated by double immunohistochemistry. Progressive renal fibrosis was associated with the loss of CD4+FOXP3+IL-17+ T cells in splenic single-cell suspensions. FOXP3+IL-17+ T cells expressed TGF-β1 both in vitro and in vivo, and TGF-β1 expression was significantly knockdown by IL-17 siRNA in vitro. These cells were found to play a role in converting Tregs into IL-17- and TGF-β1-producing cells. CONCLUSIONS TSA treatment decreased JG hyperplasia, the percentage of FOXP3+IL-17+ cells and the degree of fibrosis, suggesting that therapeutic benefits may result from epigenetic modifications.
Collapse
Affiliation(s)
- Wen-Pyng Wu
- Graduate Institute of Clinical Medical Science, College of Medicine, China Medical University, Taichung, Taiwan.,Division of Nephrology, Ching Chyuan Hospital, Taichung, Taiwan
| | - Yi-Giien Tsai
- Department of Pediatrics, Changhua Christian Hospital, Changhua, Taiwan.,School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Medicine, Chung-Shan Medical University, Taichung, Taiwan
| | - Tze-Yi Lin
- Department of pathology, China Medical University Hospital, Taichung, Taiwan
| | - Ming-Ju Wu
- School of Medicine, Chung-Shan Medical University, Taichung, Taiwan. .,Division of Nephrology, Department of Medicine, Taichung Veterans General Hospital, No. 1650, Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan, Republic of China. .,Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan. .,Graduate Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan.
| | - Ching-Yuang Lin
- Graduate Institute of Clinical Medical Science, College of Medicine, China Medical University, Taichung, Taiwan. .,Clinical Immunological Center, China Medical University Hospital, No. 2, Yude Road, Taichung, 40447, Taiwan, Republic of China.
| |
Collapse
|
20
|
Foulsham W, Marmalidou A, Amouzegar A, Coco G, Chen Y, Dana R. Review: The function of regulatory T cells at the ocular surface. Ocul Surf 2017; 15:652-659. [PMID: 28576753 DOI: 10.1016/j.jtos.2017.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/29/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022]
Abstract
Regulatory T cells (Tregs) are critical modulators of immune homeostasis. Tregs maintain peripheral tolerance to self-antigens, thereby preventing autoimmune disease. Furthermore, Tregs suppress excessive immune responses deleterious to the host. Recent research has deepened our understanding of how Tregs function at the ocular surface. This manuscript describes the classification, the immunosuppressive mechanisms, and the phenotypic plasticity of Tregs. We review the contribution of Tregs to ocular surface autoimmune disease, as well as the function of Tregs in allergy and infection at the ocular surface. Finally, we review the role of Tregs in promoting allotolerance in corneal transplantation.
Collapse
Affiliation(s)
- William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Anna Marmalidou
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Afsaneh Amouzegar
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Giulia Coco
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yihe Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Effector and Regulatory T Cell Trafficking in Corneal Allograft Rejection. Mediators Inflamm 2017; 2017:8670280. [PMID: 28539707 PMCID: PMC5429952 DOI: 10.1155/2017/8670280] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/26/2017] [Indexed: 01/08/2023] Open
Abstract
Corneal transplantation is among the most prevalent and successful forms of solid tissue transplantation in humans. Failure of corneal allograft is mainly due to immune-mediated destruction of the graft, a complex and highly coordinated process that involves elaborate interactions between cells of innate and adaptive immunity. The migration of immune cells to regional lymphoid tissues and to the site of graft plays a central role in the immunopathogenesis of graft rejection. Intricate interactions between adhesion molecules and their counter receptors on immune cells in conjunction with tissue-specific chemokines guide the trafficking of these cells to the draining lymph nodes and ultimately to the site of graft. In this review, we discuss the cascade of chemokines and adhesion molecules that mediate the trafficking of effector and regulatory T cells during corneal allograft rejection.
Collapse
|
22
|
Besançon A, Baas M, Goncalves T, Valette F, Waldmann H, Chatenoud L, You S. The Induction and Maintenance of Transplant Tolerance Engages Both Regulatory and Anergic CD4 + T cells. Front Immunol 2017; 8:218. [PMID: 28321218 PMCID: PMC5337867 DOI: 10.3389/fimmu.2017.00218] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/15/2017] [Indexed: 11/23/2022] Open
Abstract
Therapeutic tolerance to self-antigens or foreign antigens is thought to depend on constant vigilance by Foxp3+ regulatory T cells (Tregs). Previous work using a pancreatic islet allograft model and a short pulse of CD3 antibody therapy has shown that CD8+ T cells become anergic and use TGFβ and coinhibitory signaling as their contribution to the tolerance process. Here, we examine the role of CD4+ T cells in tolerization by CD3 antibodies. We show that both Foxp3+ Tregs and CD4+ T cell anergy play a role in the induction of tolerance and its maintenance. Foxp3+ Tregs resisted CD3 antibody-mediated depletion, unlike intragraft Th1 CD4+ lymphocytes coexpressing granzyme B and Tbx21, which were selectively eliminated. Tregs were mandatory for induction of tolerance as their depletion at the time of CD3 antibody therapy or for a short time thereafter, by an antibody to CD25 (PC61), led to graft rejection. Early treatment with CTLA-4 antibody gave the same outcome. In contrast, neither PC61 nor anti-CTLA-4 given late, at day 100 posttransplant, reversed tolerance once established. Ablation of Foxp3 T cells after diphtheria toxin injection in tolerant Foxp3DTR recipient mice provided the same outcome. Alloreactive T cells had been rendered intrinsically unresponsive as total CD4+ or Treg-deprived CD4+ T cells from tolerant recipients were unable to mount donor-specific IFN-γ responses. In addition, intragraft Treg-deprived CD4+ T cells lacked proliferative capacities, expressed high levels of the inhibitory receptor PD-1, and exhibited a CD73hiFR4hi phenotype, thus reflecting a state of T cell anergy. We conclude that Tregs play a substantive and critical role in guiding the immune system toward tolerance of the allograft, when induced by CD3 antibody, but are less important for maintenance of the tolerant state, where T cell anergy appears sufficient.
Collapse
Affiliation(s)
- Alix Besançon
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; INSERM U1151, Institut Necker-Enfants Malades, Paris, France; CNRS UMR 8253, Institut Necker-Enfants Malades, Paris, France
| | - Marije Baas
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; INSERM U1151, Institut Necker-Enfants Malades, Paris, France; CNRS UMR 8253, Institut Necker-Enfants Malades, Paris, France
| | - Tania Goncalves
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; INSERM U1151, Institut Necker-Enfants Malades, Paris, France; CNRS UMR 8253, Institut Necker-Enfants Malades, Paris, France
| | - Fabrice Valette
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; INSERM U1151, Institut Necker-Enfants Malades, Paris, France; CNRS UMR 8253, Institut Necker-Enfants Malades, Paris, France
| | - Herman Waldmann
- Therapeutic Immunology Group, Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| | - Lucienne Chatenoud
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; INSERM U1151, Institut Necker-Enfants Malades, Paris, France; CNRS UMR 8253, Institut Necker-Enfants Malades, Paris, France
| | - Sylvaine You
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; INSERM U1151, Institut Necker-Enfants Malades, Paris, France; CNRS UMR 8253, Institut Necker-Enfants Malades, Paris, France
| |
Collapse
|
23
|
Khan MA. T regulatory cell mediated immunotherapy for solid organ transplantation: A clinical perspective. Mol Med 2017; 22:892-904. [PMID: 27878210 PMCID: PMC5319206 DOI: 10.2119/molmed.2016.00050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 11/11/2016] [Indexed: 12/12/2022] Open
Abstract
T regulatory cells (Tregs) play a vital role in suppressing heightened immune responses, and thereby promote a state of immunological tolerance. Tregs modulate both innate and adaptive immunity, which make them a potential candidate for cell-based immunotherapy to suppress uncontrolled activation of graft specific inflammatory cells and their toxic mediators. These grafts specific inflammatory cells (T effector cells) and other inflammatory mediators (Immunoglobulins, active complement mediators) are mainly responsible for graft vascular deterioration followed by acute/chronic rejection. Treg mediated immunotherapy is under investigation to induce allospecific tolerance in various ongoing clinical trials in organ transplant recipients. Treg immunotherapy is showing promising results but the key issues regarding Treg immunotherapy are not yet fully resolved including their mechanism of action, and specific Treg cell phenotype responsible for a state of tolerance. This review highlights the involvement of various subsets of Tregs during immune suppression, novelty of Tregs functions, effects on angiogenesis, emerging technologies for effective Treg expansion, plasticity and safety associated with clinical applications. Altogether this information will assist in designing single/combined Treg mediated therapies for successful clinical trials in solid organ transplantations.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia 11211
| |
Collapse
|
24
|
Chang J, Graves SS, Butts-Miwongtum T, Sale GE, Storb R, Mathes DW. Long-term Tolerance Toward Haploidentical Vascularized Composite Allograft Transplantation in a Canine Model Using Bone Marrow or Mobilized Stem Cells. Transplantation 2016; 100:e120-e127. [PMID: 27861292 PMCID: PMC5453180 DOI: 10.1097/tp.0000000000001496] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND The development of safe and reliable protocols for the transplantation of the face and hands may be accomplished with animal modeling of transplantation of vascularized composite allografts (VCA). Previously, we demonstrated that tolerance to a VCA could be achieved after canine recipients were simultaneously given marrow from a dog leukocyte antigen-identical donor. In the present study, we extend those findings across a dog leukocyte antigen mismatched barrier. METHODS Eight recipient dogs received total body irradiation (4.5 cGy), hematopoietic cell transplantation (HCT), either marrow (n = 4) or granulocyte-colony stimulating factor mobilized peripheral blood stem cells (n = 4), and a VCA transplant from the HCT donor. Post grafting immunosuppression consisted of mycophenolate mofetil (28 days) and cyclosporine (35 days). RESULTS In 4 dogs receiving bone marrow, 1 accepted both its marrow transplant and demonstrated long-term tolerance to the donor VCA (>52 weeks). Three dogs rejected both their marrow transplants and VCA at 5 to 7 weeks posttransplant. Dogs receiving mobilized stem cells all accepted their stem cell transplant and became tolerant to the VCA. However, 3 dogs developed graft-versus-host disease, whereas 1 dog rejected its stem cell graft by week 15 but exhibited long-term tolerance toward its VCA (>90 weeks). CONCLUSIONS The data suggest that simultaneous transplantation of mobilized stem cells and a VCA is feasible and leads to tolerance toward the VCA in a haploidentical setting. However, there is a higher rate of donor stem cell engraftment compared with marrow HCT and an increase in the incidence of graft-versus-host disease.
Collapse
Affiliation(s)
- Jeff Chang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Surgery, University of Washington, Seattle, WA
| | - Scott S. Graves
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | | | - George E. Sale
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Pathology, University of Washington, Seattle, WA
| | - Rainer Storb
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - David W. Mathes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Surgery, University of Colorado, Aurora, CO
- Plastic Surgery Service, VA Eastern Colorado Care System, Denver, CO
| |
Collapse
|
25
|
A novel differentiation pathway from CD4⁺ T cells to CD4⁻ T cells for maintaining immune system homeostasis. Cell Death Dis 2016; 7:e2193. [PMID: 27077809 PMCID: PMC4855662 DOI: 10.1038/cddis.2016.83] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/07/2016] [Accepted: 03/09/2016] [Indexed: 12/26/2022]
Abstract
CD4+ T lymphocytes are key players in the adaptive immune system and can differentiate into a variety of effector and regulatory T cells. Here, we provide evidence that a novel differentiation pathway of CD4+ T cells shifts the balance from a destructive T-cell response to one that favors regulation in an immune-mediated liver injury model. Peripheral CD4−CD8−NK1.1− double-negative T cells (DNT) was increased following Concanavalin A administration in mice. Adoptive transfer of DNT led to significant protection from hepatocyte necrosis by direct inhibition on the activation of lymphocytes, a process that occurred primarily through the perforin-granzyme B route. These DNT converted from CD4+ rather than CD8+ T cells, a process primarily regulated by OX40. DNT migrated to the liver through the CXCR3-CXCL9/CXCL10 interaction. In conclusion, we elucidated a novel differentiation pathway from activated CD4+ T cells to regulatory DNT cells for maintaining homeostasis of the immune system in vivo, and provided key evidence that utilizing this novel differentiation pathway has potential application in the prevention and treatment of autoimmune diseases.
Collapse
|
26
|
You S. Differential sensitivity of regulatory and effector T cells to cell death: a prerequisite for transplant tolerance. Front Immunol 2015; 6:242. [PMID: 26042125 PMCID: PMC4437185 DOI: 10.3389/fimmu.2015.00242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/06/2015] [Indexed: 12/13/2022] Open
Abstract
Despite significant progress achieved in transplantation, immunosuppressive therapies currently used to prevent graft rejection are still endowed with severe side effects impairing their efficiency over the long term. Thus, the development of graft-specific, non-toxic innovative therapeutic strategies has become a major challenge, the goal being to selectively target alloreactive effector T cells while sparing CD4+Foxp3+ regulatory T cells (Tregs) to promote operational tolerance. Various approaches, notably the one based on monoclonal antibodies or fusion proteins directed against the TCR/CD3 complex, TCR coreceptors, or costimulatory molecules, have been proposed to reduce the alloreactive T cell pool, which is an essential prerequisite to create a therapeutic window allowing Tregs to induce and maintain allograft tolerance. In this mini review, we focus on the differential sensitivity of Tregs and effector T cells to the depleting and inhibitory effect of these immunotherapies, with a particular emphasis on CD3-specific antibodies that beyond their immunosuppressive effect, also express potent tolerogenic capacities.
Collapse
Affiliation(s)
- Sylvaine You
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; INSERM U1151, Institut Necker-Enfants Malades , Paris , France ; CNRS UMR 8253, Institut Necker-Enfants Malades , Paris , France
| |
Collapse
|
27
|
Guo Y, Brown C, Ortiz C, Noelle RJ. Leukocyte homing, fate, and function are controlled by retinoic acid. Physiol Rev 2015; 95:125-48. [PMID: 25540140 DOI: 10.1152/physrev.00032.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Although vitamin A was recognized as an "anti-infective vitamin" over 90 years ago, the mechanism of how vitamin A regulates immunity is only beginning to be understood. Early studies which focused on the immune responses in vitamin A-deficient (VAD) animals clearly demonstrated compromised immunity and consequently increased susceptibility to infectious disease. The active form of vitamin A, retinoic acid (RA), has been shown to have a profound impact on the homing and differentiation of leukocytes. Both pharmacological and genetic approaches have been applied to the understanding of how RA regulates the development and differentiation of various immune cell subsets, and how RA influences the development of immunity versus tolerance. These studies clearly show that RA profoundly impacts on cell- and humoral-mediated immunity. In this review, the early findings on the complex relationship between VAD and immunity are discussed as well as vitamin A metabolism and signaling within hematopoietic cells. Particular attention is focused on how RA impacts on T-cell lineage commitment and plasticity in various diseases.
Collapse
Affiliation(s)
- Yanxia Guo
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, New Hampshire; and Medical Research Council Centre of Transplantation, Guy's Hospital, King's College London, King's Health Partners, London, United Kingdom
| | - Chrysothemis Brown
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, New Hampshire; and Medical Research Council Centre of Transplantation, Guy's Hospital, King's College London, King's Health Partners, London, United Kingdom
| | - Carla Ortiz
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, New Hampshire; and Medical Research Council Centre of Transplantation, Guy's Hospital, King's College London, King's Health Partners, London, United Kingdom
| | - Randolph J Noelle
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, New Hampshire; and Medical Research Council Centre of Transplantation, Guy's Hospital, King's College London, King's Health Partners, London, United Kingdom
| |
Collapse
|
28
|
Donor CD4+ Foxp3+ regulatory T cells are necessary for posttransplantation cyclophosphamide-mediated protection against GVHD in mice. Blood 2014; 124:2131-41. [PMID: 25139358 DOI: 10.1182/blood-2013-10-525873] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Posttransplantation cyclophosphamide (PTCy) is an effective prophylaxis against graft-versus-host disease (GVHD). However, it is unknown whether PTCy works singularly by eliminating alloreactive T cells via DNA alkylation or also by restoring the conventional (Tcon)/regulatory (Treg) T-cell balance. We studied the role of Tregs in PTCy-mediated GVHD prophylaxis in murine models of allogeneic blood or marrow transplantation (alloBMT). In 2 distinct MHC-matched alloBMT models, infusing Treg-depleted allografts abrogated the GVHD-prophylactic activity of PTCy. Using allografts in which Foxp3(+) Tregs could be selectively depleted in vivo, either pre- or post-PTCy ablation of donor thymus-derived Tregs (tTregs) abolished PTCy protection against GVHD. PTCy treatment was associated with relative preservation of donor Tregs. Experiments using combinations of Foxp3(-) Tcons and Foxp3(+) Tregs sorted from different Foxp3 reporter mice indicated that donor Treg persistence after PTCy treatment was predominantly caused by survival of functional tTregs that retained Treg-specific demethylation and also induction of peripherally derived Tregs. Finally, adoptive transfer of tTregs retrieved from PTCy-treated chimeras rescued PTCy-treated, Treg-depleted recipients from lethal GVHD. Our findings indicate that PTCy-mediated protection against GVHD is not singularly dependent on depletion of donor alloreactive T cells but also requires rapidly recovering donor Tregs to initiate and maintain alloimmune regulation.
Collapse
|
29
|
Krupnick AS, Lin X, Li W, Higashikubo R, Zinselmeyer BH, Hartzler H, Toth K, Ritter JH, Berezin MY, Wang ST, Miller MJ, Gelman AE, Kreisel D. Central memory CD8+ T lymphocytes mediate lung allograft acceptance. J Clin Invest 2014; 124:1130-43. [PMID: 24569377 PMCID: PMC3938255 DOI: 10.1172/jci71359] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 12/05/2013] [Indexed: 12/31/2022] Open
Abstract
Memory T lymphocytes are commonly viewed as a major barrier for long-term survival of organ allografts and are thought to accelerate rejection responses due to their rapid infiltration into allografts, low threshold for activation, and ability to produce inflammatory mediators. Because memory T cells are usually associated with rejection, preclinical protocols have been developed to target this population in transplant recipients. Here, using a murine model, we found that costimulatory blockade-mediated lung allograft acceptance depended on the rapid infiltration of the graft by central memory CD8+ T cells (CD44(hi)CD62L(hi)CCR7+). Chemokine receptor signaling and alloantigen recognition were required for trafficking of these memory T cells to lung allografts. Intravital 2-photon imaging revealed that CCR7 expression on CD8+ T cells was critical for formation of stable synapses with antigen-presenting cells, resulting in IFN-γ production, which induced NO and downregulated alloimmune responses. Thus, we describe a critical role for CD8+ central memory T cells in lung allograft acceptance and highlight the need for tailored approaches for tolerance induction in the lung.
Collapse
Affiliation(s)
- Alexander Sasha Krupnick
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Xue Lin
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Wenjun Li
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ryuiji Higashikubo
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bernd H. Zinselmeyer
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Hollyce Hartzler
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kelsey Toth
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jon H. Ritter
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mikhail Y. Berezin
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Steven T. Wang
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mark J. Miller
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Andrew E. Gelman
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Daniel Kreisel
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
30
|
Amelioration of ovalbumin-induced allergic airway disease following Der p 1 peptide immunotherapy is not associated with induction of IL-35. Mucosal Immunol 2014; 7:379-90. [PMID: 23945544 DOI: 10.1038/mi.2013.56] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 07/01/2013] [Indexed: 02/04/2023]
Abstract
In the present study, we show therapeutic amelioration of established ovalbumin (OVA)-induced allergic airway disease following house dust mite (HDM) peptide therapy. Mice were sensitized and challenged with OVA and HDM protein extract (Dermatophagoides species) to induce dual allergen sensitization and allergic airway disease. Treatment of allergic mice with peptides derived from the major allergen Der p 1 suppressed OVA-induced airway hyperresponsiveness, tissue eosinophilia, and goblet cell hyperplasia upon rechallenge with allergen. Peptide treatment also suppressed OVA-specific T-cell proliferation. Resolution of airway pathophysiology was associated with a reduction in recruitment, proliferation, and effector function of T(H)2 cells and decreased interleukin (IL)-17⁺ T cells. Furthermore, peptide immunotherapy induced the regulatory cytokine IL-10 and increased the proportion of Fox p3⁺ cells among those expressing IL-10. Tolerance to OVA was not associated with increased IL-35. In conclusion, our results provide in vivo evidence for the creation of a tolerogenic environment following HDM peptide immunotherapy, leading to the therapeutic amelioration of established OVA-induced allergic airway disease.
Collapse
|
31
|
Lee K, Nguyen V, Lee KM, Kang SM, Tang Q. Attenuation of donor-reactive T cells allows effective control of allograft rejection using regulatory T cell therapy. Am J Transplant 2014; 14:27-38. [PMID: 24354870 PMCID: PMC5262439 DOI: 10.1111/ajt.12509] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 07/01/2013] [Accepted: 08/01/2013] [Indexed: 01/25/2023]
Abstract
Regulatory T cells (Tregs) are essential for the establishment and maintenance of immune tolerance, suggesting a potential therapeutic role for Tregs in transplantation. However, Treg administration alone is insufficient in inducing long-term allograft survival in normal hosts, likely due to the high frequency of alloreactive T cells. We hypothesized that a targeted reduction of alloreactive T effector cells would allow a therapeutic window for Treg efficacy. Here we show that preconditioning recipient mice with donor-specific transfusion followed by cyclophosphamide treatment deleted 70-80% donor-reactive T cells, but failed to prolong islet allograft survival. However, infusion of either 5 × 10(6) Tregs with direct donor reactivity or 25 × 10(6) polyclonal Tregs led to indefinite survival of BALB/c islets in more than 70% of preconditioned C57BL/6 recipients. Notably, protection of C3H islets in autoimmune nonobese diabetic mice required islet autoantigen-specific Tregs together with polyclonal Tregs. Treg therapy led to significant reduction of CD8(+) T cells and concomitant increase in endogenous Tregs among graft-infiltrating cells early after transplantation. Together, these results demonstrate that reduction of the donor-reactive T cells will be an important component of Treg-based therapies in transplantation.
Collapse
Affiliation(s)
- K. Lee
- Department of Surgery, University of California, San Francisco, San Francisco, CA,Department of Biochemistry and Molecular Biology, Korea University, Seoul, Republic of Korea
| | - V. Nguyen
- Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - K.-M. Lee
- Department of Biochemistry and Molecular Biology, Korea University, Seoul, Republic of Korea
| | - S.-M. Kang
- Department of Surgery, University of California, San Francisco, San Francisco, CA,Corresponding authors: Qizhi Tang, and Sang-Mo Kang,
| | - Q. Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA,Corresponding authors: Qizhi Tang, and Sang-Mo Kang,
| |
Collapse
|
32
|
Chauhan SK, Saban DR, Dohlman TH, Dana R. CCL-21 conditioned regulatory T cells induce allotolerance through enhanced homing to lymphoid tissue. THE JOURNAL OF IMMUNOLOGY 2013; 192:817-23. [PMID: 24337379 DOI: 10.4049/jimmunol.1203469] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Regulatory T cells (Tregs) are instrumental in the induction and maintenance of tolerance, including in transplantation. Tregs induce allotolerance by interacting with APCs and T cells, interactions that require their proper homing to the lymphoid tissues. Using a well-characterized model of corneal allotransplantation, we demonstrate in this study that Tregs in the draining lymph nodes (LN) of allograft acceptors, but not rejectors, colocalize with APCs in the paracortical areas and express high levels of CCR7. In addition, we show that Treg expression of CCR7 is important not only for Treg homing to the draining LN, but also for optimal Treg suppressive function. Finally, we show that Tregs augmented for CCR7 expression by their ex vivo stimulation with the CCR7 ligand CCL21 show enhanced homing to the draining LN of allograft recipients and promote transplant survival. Together, these findings suggest that CCR7 expression is critical for Treg function and migration and that conditioning of Treg for maximal CCR7 expression may be a viable strategy for promoting allograft survival.
Collapse
Affiliation(s)
- Sunil K Chauhan
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02144
| | | | | | | |
Collapse
|
33
|
Putnam AL, Safinia N, Medvec A, Laszkowska M, Wray M, Mintz MA, Trotta E, Szot GL, Liu W, Lares A, Lee K, Laing A, Lechler RI, Riley JL, Bluestone JA, Lombardi G, Tang Q. Clinical grade manufacturing of human alloantigen-reactive regulatory T cells for use in transplantation. Am J Transplant 2013; 13:3010-20. [PMID: 24102808 PMCID: PMC4161737 DOI: 10.1111/ajt.12433] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/15/2013] [Accepted: 07/15/2013] [Indexed: 01/25/2023]
Abstract
Regulatory T cell (Treg) therapy has the potential to induce transplantation tolerance so that immunosuppression and associated morbidity can be minimized. Alloantigen-reactive Tregs (arTregs) are more effective at preventing graft rejection than polyclonally expanded Tregs (PolyTregs) in murine models. We have developed a manufacturing process to expand human arTregs in short-term cultures using good manufacturing practice-compliant reagents. This process uses CD40L-activated allogeneic B cells to selectively expand arTregs followed by polyclonal restimulation to increase yield. Tregs expanded 100- to 1600-fold were highly alloantigen reactive and expressed the phenotype of stable Tregs. The alloantigen-expanded Tregs had a diverse TCR repertoire. They were more potent than PolyTregs in vitro and more effective at controlling allograft injuries in vivo in a humanized mouse model.
Collapse
Affiliation(s)
- A. L. Putnam
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA
| | - N. Safinia
- MRC Centre for Transplantation, King’s College London, London, UK
| | - A. Medvec
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | - M. Laszkowska
- Department of Surgery, University of California, San Francisco, San Francisco CA
| | - M. Wray
- Department of Surgery, University of California, San Francisco, San Francisco CA
| | - M. A. Mintz
- Department of Surgery, University of California, San Francisco, San Francisco CA
| | - E. Trotta
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA,Department of Surgery, University of California, San Francisco, San Francisco CA
| | - G. L. Szot
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA,Department of Surgery, University of California, San Francisco, San Francisco CA
| | - W. Liu
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA
| | - A. Lares
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA
| | - K. Lee
- Department of Surgery, University of California, San Francisco, San Francisco CA
| | - A. Laing
- MRC Centre for Transplantation, King’s College London, London, UK
| | - R. I. Lechler
- MRC Centre for Transplantation, King’s College London, London, UK
| | - J. L. Riley
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | - J. A. Bluestone
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA
| | - G. Lombardi
- MRC Centre for Transplantation, King’s College London, London, UK
| | - Q. Tang
- Department of Surgery, University of California, San Francisco, San Francisco CA,Corresponding author: Qizhi Tang,
| |
Collapse
|
34
|
Dons E, Raimondi G, Zhang H, Zahorchak A, Bhama J, Lu L, Ezzelarab M, Ijzermans J, Cooper D, Thomson A. Ex vivo-expanded cynomolgus macaque regulatory T cells are resistant to alemtuzumab-mediated cytotoxicity. Am J Transplant 2013; 13:2169-78. [PMID: 23635093 PMCID: PMC3780590 DOI: 10.1111/ajt.12248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 03/07/2013] [Accepted: 03/10/2013] [Indexed: 01/25/2023]
Abstract
Alemtuzumab (Campath-1H) is a humanized monoclonal antibody (Ab) directed against CD52 that depletes lymphocytes and other leukocytes, mainly by complement-dependent mechanisms. We investigated the influence of alemtuzumab (i) on ex vivo-expanded cynomolgus monkey regulatory T cells (Treg) generated for prospective use in adoptive cell therapy and (ii) on naturally occurring Treg following alemtuzumab infusion. Treg were isolated from PBMC and lymph nodes and expanded for two rounds. CD52 expression, binding of alemtuzumab and both complement-mediated killing and Ab-dependent cell-mediated cytotoxicity (ADCC) were compared between freshly isolated and expanded Treg and effector T cells. Monkeys undergoing allogeneic heart transplantation given alemtuzumab were monitored for Treg and serum alemtuzumab activity. Ex vivo-expanded Treg showed progressive downregulation of CD52 expression, absence of alemtuzumab binding, minimal change in complement inhibitory protein (CD46) expression and no complement-dependent killing or ADCC. Infusion of alemtuzumab caused potent depletion of all lymphocytes, but a transient increase in the incidence of circulating Treg. After infusion of alemtuzumab, monkey serum killed fresh PBMC, but not expanded Treg. Thus, expanded cynomolgus monkey Treg are resistant to alemtuzumab-mediated, complement-dependent cytotoxicity. Furthermore, our data suggest that these expanded monkey Treg can be infused into graft recipients given alemtuzumab without risk of complement-mediated killing.
Collapse
Affiliation(s)
- E.M. Dons
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA,Department of Surgery, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - G. Raimondi
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - H. Zhang
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - A.F. Zahorchak
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - J.K. Bhama
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center Presbyterian, Pittsburgh, PA
| | - L. Lu
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - M. Ezzelarab
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - J.N.M. Ijzermans
- Department of Surgery, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - D.K.C. Cooper
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - A.W. Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA,Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
35
|
Commodaro AG, Pedregosa JF, Peron JP, Brandão W, Rizzo LV, Bueno V. The imbalance between Treg and Th17 cells caused by FTY720 treatment in skin allograft rejection. Clinics (Sao Paulo) 2012; 67:805-13. [PMID: 22892927 PMCID: PMC3400173 DOI: 10.6061/clinics/2012(07)17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 03/14/2012] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES FTY720 modulates CD4+T cells by the augmentation of regulatory T cell activity, secretion of suppressive cytokines and suppression of IL-17 secretion by Th17 cells. To further understand the process of graft rejection/acceptance, we evaluated skin allograft survival and associated events after FTY720 treatment. METHODS F1 mice (C57BL/6xBALB/c) and C57BL/6 mice were used as donors for and recipients of skin transplantation, respectively. The recipients were transplanted and either not treated or treated with FTY720 by gavage for 21 days to evaluate the allograft survival. In another set of experiments, the immunological evaluation was performed five days post-transplantation. The spleens, axillary lymph nodes and skin allografts of the recipient mice were harvested for phenotyping (flow cytometry), gene expression (real-time PCR) and cytokine (Bio-Plex) analysis. RESULTS The FTY720 treatment significantly increased skin allograft survival, reduced the number of cells in the lymph nodes and decreased the percentage of Tregs at this site in the C57BL/6 recipients. Moreover, the treatment reduced the number of graft-infiltrating cells and the percentage of CD4+ graft-infiltrating cells. The cytokine analysis (splenocytes) showed decreased levels of IL-10, IL-6 and IL-17 in the FTY720-treated mice. We also observed a decrease in the IL-10, IL-6 and IL-23 mRNA levels, as well as an increase in the IL-27 mRNA levels, in the splenocytes of the treated group. The FTY720-treated mice exhibited increased mRNA levels of IL-10, IL-27 and IL-23 in the skin graft. CONCLUSIONS Our results demonstrated prolonged but not indefinite skin allograft survival by FTY720 treatment. This finding indicates that the drug did not prevent the imbalance between Tr1 and Th17 cells in the graft that led to rejection.
Collapse
|
36
|
Perz JB, Gürel S, Schonland SO, Hegenbart U, Ho AD, Dreger P. CD4+CD25highCD127low regulatory T cells in peripheral blood are not an independent factor for chronic graft-versus-host disease after allogeneic stem cell transplantation. ScientificWorldJournal 2012; 2012:606839. [PMID: 22666141 PMCID: PMC3361289 DOI: 10.1100/2012/606839] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Accepted: 11/30/2011] [Indexed: 11/17/2022] Open
Abstract
Background. The therapeutic efficacy of allogeneic hemopoietic stem cell transplantation (HSCT) largely relies on the graft-versus-leukemia (GVL) effect. Uncontrolled graft-versus-host disease (GVHD) is a feared complication of HSCT. Regulatory T cells (Treg) are a subset of CD4+ T-helper cells believed to maintain tolerance after HSCT. It remains unclear whether low peripheral blood Treg have an impact on the risk for acute (aGVHD) and chronic GVHD (cGVHD). Methods. In this paper we enumerated the CD4+CD25highCD127low Treg in the peripheral blood of 84 patients after at least 150 days from HSCT and in 20 healthy age-matched controls. Results. Although similar mean lymphocyte counts were found in patients and controls, CD3+CD4+ T-cell counts were significantly lower in patients. Patients also had significantly lower Treg percentages among lymphocytes as compared to controls. Patients with cGVHD had even higher percentages of Treg if compared to patients without cGVHD. In multivariate analysis, Treg percentages were not an independent factor for cGVHD. Conclusions. This paper did not show a relation between deficient peripheral blood Treg and cGVHD, therefore cGVHD does not seem to occur as a result of peripheral Treg paucity.
Collapse
Affiliation(s)
- Jolanta B Perz
- Department of Internal Medicine-Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Regulatory T cells (Tregs) are long-lived cells that suppress immune responses in vivo in a dominant and antigen-specific manner. Therefore, therapeutic application of Tregs to control unwanted immune responses is an active area of investigation. Tregs can confer long-term protection against auto-inflammatory diseases in mouse models. They have also been shown to be effective in suppressing alloimmunity in models of graft-versus-host disease and organ transplantation. Building on extensive research in Treg biology and preclinical testing of therapeutic efficacy over the past decade, we are now at the point of evaluating the safety and efficacy of Treg therapy in humans. This review focuses on developing therapy for transplantation using CD4(+)Foxp3(+) Tregs, with an emphasis on the studies that have informed clinical approaches that aim to maximize the benefits while overcoming the challenges and risks of Treg cell therapy.
Collapse
Affiliation(s)
- Qizhi Tang
- Division of Transplantation, Department of Surgery, University of California San Francisco, San Francisco, CA 94143-0780, USA.
| | | | | |
Collapse
|
38
|
Bhatt S, Fung JJ, Lu L, Qian S. Tolerance-inducing strategies in islet transplantation. Int J Endocrinol 2012; 2012:396524. [PMID: 22675353 PMCID: PMC3366204 DOI: 10.1155/2012/396524] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 03/08/2012] [Indexed: 12/12/2022] Open
Abstract
Allogeneic islet transplantation is a promising approach for restoring normoglycemia in type 1 diabetic patients. Current use of immunosuppressive therapies for management of islet transplant recipients can be counterintuitive to islet function and can lead to complications in the long term. The induction of donor-specific tolerance eliminates the dependency on immunosuppression and allows recipients to retain responses to foreign antigens. The mechanisms by which tolerance is achieved involve the deletion of donor-reactive T cells, induction of T-cell anergy, immune deviation, and generation of regulatory T cells. This review will outline the various methods used for inducing donor-specific tolerance in islet transplantation and will highlight the previously unforeseen potential of tissue stromal cells in promoting islet engraftment.
Collapse
Affiliation(s)
- Sumantha Bhatt
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - John J. Fung
- Department of General Surgery, Transplant Center, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lina Lu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of General Surgery, Transplant Center, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shiguang Qian
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of General Surgery, Transplant Center, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- *Shiguang Qian:
| |
Collapse
|
39
|
Gagliani N, Gregori S, Jofra T, Valle A, Stabilini A, Rothstein DM, Atkinson M, Roncarolo MG, Battaglia M. Rapamycin combined with anti-CD45RB mAb and IL-10 or with G-CSF induces tolerance in a stringent mouse model of islet transplantation. PLoS One 2011; 6:e28434. [PMID: 22174806 PMCID: PMC3235119 DOI: 10.1371/journal.pone.0028434] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 11/08/2011] [Indexed: 02/07/2023] Open
Abstract
Background A large pool of preexisting alloreactive effector T cells can cause allogeneic graft rejection following transplantation. However, it is possible to induce transplant tolerance by altering the balance between effector and regulatory T (Treg) cells. Among the various Treg-cell types, Foxp3+Treg and IL-10–producing T regulatory type 1 (Tr1) cells have frequently been associated with tolerance following transplantation in both mice and humans. Previously, we demonstrated that rapamycin+IL-10 promotes Tr1-cell–associated tolerance in Balb/c mice transplanted with C57BL/6 pancreatic islets. However, this same treatment was unsuccessful in C57BL/6 mice transplanted with Balb/c islets (classified as a stringent transplant model). We accordingly designed a protocol that would be effective in the latter transplant model by simultaneously depleting effector T cells and fostering production of Treg cells. We additionally developed and tested a clinically translatable protocol that used no depleting agent. Methodology/Principal Findings Diabetic C57BL/6 mice were transplanted with Balb/c pancreatic islets. Recipient mice transiently treated with anti-CD45RB mAb+rapamycin+IL-10 developed antigen-specific tolerance. During treatment, Foxp3+Treg cells were momentarily enriched in the blood, followed by accumulation in the graft and draining lymph node, whereas CD4+IL-10+IL-4− T (i.e., Tr1) cells localized in the spleen. In long-term tolerant mice, only CD4+IL-10+IL-4− T cells remained enriched in the spleen and IL-10 was key in the maintenance of tolerance. Alternatively, recipient mice were treated with two compounds routinely used in the clinic (namely, rapamycin and G-CSF); this drug combination promoted tolerance associated with CD4+IL-10+IL-4− T cells. Conclusions/Significance The anti-CD45RB mAb+rapamycin+IL-10 combined protocol promotes a state of tolerance that is IL-10 dependent. Moreover, the combination of rapamycin+G-CSF induces tolerance and such treatment could be readily translatable into the clinic.
Collapse
Affiliation(s)
- Nicola Gagliani
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Tatiana Jofra
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Valle
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Angela Stabilini
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | - David M. Rothstein
- Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Mark Atkinson
- Department of Pathology, The University of Florida, Gainesville, Florida, United States of America
| | - Maria Grazia Roncarolo
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Manuela Battaglia
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- * E-mail:
| |
Collapse
|
40
|
B7-H4 Pathway in Islet Transplantation and β-Cell Replacement Therapies. J Transplant 2011; 2011:418902. [PMID: 22028949 PMCID: PMC3196026 DOI: 10.1155/2011/418902] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 08/03/2011] [Indexed: 12/18/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease and characterized by absolute insulin deficiency. β-cell replacement by islet cell transplantation has been established as a feasible treatment option for T1D. The two main obstacles after islet transplantation are alloreactive T-cell-mediated graft rejection and recurrence of autoimmune diabetes mellitus in recipients. T cells play a central role in determining the outcome of both autoimmune responses and allograft survival. B7-H4, a newly identified B7 homolog, plays a key role in maintaining T-cell homeostasis by reducing T-cell proliferation and cytokine production. The relationship between B7-H4 and allograft survival/autoimmunity has been investigated recently in both islet transplantation and the nonobese diabetic (NOD) mouse models. B7-H4 protects allograft survival and generates donor-specific tolerance. It also prevents the development of autoimmune diabetes. More importantly, B7-H4 plays an indispensable role in alloimmunity in the absence of the classic CD28/CTLA-4 : B7 pathway, suggesting a synergistic/additive effect with other agents such as CTLA-4 on inhibition of unwanted immune responses.
Collapse
|
41
|
Dodd-o JM, Lendermon EA, Miller HL, Zhong Q, John ER, Jungraithmayr WM, D'Alessio FR, McDyer JF. CD154 blockade abrogates allospecific responses and enhances CD4(+) regulatory T-cells in mouse orthotopic lung transplant. Am J Transplant 2011; 11:1815-24. [PMID: 21827610 PMCID: PMC3827913 DOI: 10.1111/j.1600-6143.2011.03623.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Acute cellular rejection (ACR) is a common and important clinical complication following lung transplantation. While there is a clinical need for the development of novel therapies to prevent ACR, the regulation of allospecific effector T-cells in this process remains incompletely understood. Using the MHC-mismatched mouse orthotopic lung transplant model, we investigated the short-term role of anti-CD154 mAb therapy alone on allograft pathology and alloimmune T-cell effector responses. Untreated C57BL/6 recipients of BALB/c left lung allografts had high-grade rejection and diminished CD4(+) : CD8(+) graft ratios, marked by predominantly CD8(+) >CD4(+) IFN-γ(+) allospecific effector responses at day 10, compared to isograft controls. Anti-CD154 mAb therapy strikingly abrogated both CD8(+) and CD4(+) alloeffector responses and significantly increased lung allograft CD4(+) : CD8(+) ratios. Examination of graft CD4(+) T-cells revealed significantly increased frequencies of CD4(+) CD25(+) Foxp3(+) regulatory T-cells in the lung allografts of anti-CD154-treated mice and was associated with significant attenuation of ACR compared to untreated controls. Together, these data show that CD154/CD40 costimulation blockade alone is sufficient to abrogate allospecific effector T-cell responses and significantly shifts the lung allograft toward an environment predominated by CD4(+) T regulatory cells in association with an attenuation of ACR.
Collapse
Affiliation(s)
- J M Dodd-o
- Department of Anesthesiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Seung E, Cho JL, Sparwasser T, Medoff BD, Luster AD. Inhibiting CXCR3-dependent CD8+ T cell trafficking enhances tolerance induction in a mouse model of lung rejection. THE JOURNAL OF IMMUNOLOGY 2011; 186:6830-8. [PMID: 21555535 DOI: 10.4049/jimmunol.1001049] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Lung transplantation remains the only effective therapy for patients with end-stage pulmonary diseases. Unfortunately, acute rejection of the lung remains a frequent complication and is an important cause of morbidity and mortality. The induction of transplant tolerance is thought to be dependent, in part, on the balance between allograft effector mechanisms mediated by effector T lymphocytes (Teff), and regulatory mechanisms mediated by FOXP3(+) regulatory T cells (Treg). In this study, we explored an approach to tip the balance in favor of regulatory mechanisms by modulating chemokine activity. We demonstrate in an adoptive transfer model of lung rejection that CXCR3-deficient CD8(+) Teff have impaired migration into the lungs compared with wild-type Teff, which results in a dramatic reduction in fatal pulmonary inflammation. The lungs of surviving mice contained tolerized CXCR3-deficient Teff, as well as a large increase in Treg. We confirmed that Treg were needed for tolerance and that their ability to induce tolerance was dependent on their numbers in the lung relative to the numbers of Teff. These data suggest that transplantation tolerance can be achieved by reducing the recruitment of some, but not necessarily all, CD8(+) Teff into the target organ and suggest a novel approach to achieve transplant tolerance.
Collapse
Affiliation(s)
- Edward Seung
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | |
Collapse
|
43
|
Bozulic LD, Wen Y, Xu H, Ildstad ST. Evidence that FoxP3+ regulatory T cells may play a role in promoting long-term acceptance of composite tissue allotransplants. Transplantation 2011; 91:908-15. [PMID: 21304439 PMCID: PMC3592205 DOI: 10.1097/tp.0b013e31820fafb4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND FoxP3/CD4/CD25 regulatory T cells (Treg) play an important role in maintaining peripheral tolerance and are potent suppressors of T-cell activation. In this study, we evaluated the role of Treg in peripheral tolerance to composite tissue allografts (CTA). METHODS Mixed allogeneic chimeric rats were prepared by preconditioning recipients with anti-αβ-T-cell receptor monoclonal antibody followed by total body irradiation. Animals received T-cell-depleted August Copenhagen Irish bone marrow cells followed by antilymphocyte serum and FK-506. A modified osteomyocutaneous hindlimb flap composed of bone and all limb tissue components was placed in animals with chimerism greater than or equal to 1% on day 28. Recipients with CTA surviving more than or equal to 6 months were evaluated for Treg. Skin samples from tolerant long-term allogeneic transplanted, syngeneic transplanted, rejected, and naïve animals were immunostained with fluorochrome-conjugated anti-FoxP3 and anti-CD4 monoclonal antibody and visualized under a laser confocal microscope. RESULTS Significant CD4/FoxP3 Treg infiltrates were observed in tolerant donor-allograft skin samples. No graft infiltrating FoxP3 cells were observed in rejector, naïve, or skin from syngeneic CTA. In parallel experiments, mixed leukocyte reaction assays were performed to investigate the suppressor function of Treg cells. Splenocytes from tolerant, rejected, and naïve rats were sorted by flow cytometry for CD4/CD25 T cells. Treg demonstrated similar suppressive levels between the three groups. CONCLUSIONS These data suggest that Treg may play an important role in maintenance of tolerance and promoting graft acceptance in long-term CTA acceptors and may explain the favorable outcomes observed in clinical CTA recipients.
Collapse
Affiliation(s)
- Larry D. Bozulic
- Regenerex, LLC, 333 East Main Street, Suite 400, Louisville, KY 40202
| | - Yujie Wen
- Institute for Cellular Therapeutics, University of Louisville, 570 S. Preston Street, Suite 404, Louisville, KY 40202-1760
| | - Hong Xu
- Institute for Cellular Therapeutics, University of Louisville, 570 S. Preston Street, Suite 404, Louisville, KY 40202-1760
| | - Suzanne T. Ildstad
- Institute for Cellular Therapeutics, University of Louisville, 570 S. Preston Street, Suite 404, Louisville, KY 40202-1760
| |
Collapse
|
44
|
Booth AJ, Bishop DK. TGF-beta, IL-6, IL-17 and CTGF direct multiple pathologies of chronic cardiac allograft rejection. Immunotherapy 2010; 2:511-20. [PMID: 20636005 DOI: 10.2217/imt.10.33] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cardiac transplantation is an effective treatment for heart failure refractive to therapy. Although immunosuppressive therapeutics have increased first year survival rates, chronic rejection remains a significant barrier to long-term graft survival. Chronic rejection manifests as patchy interstitial fibrosis, vascular occlusion and progressive loss of graft function. Recent evidence from experimental and patient studies suggests that the development of cardiomyocyte hypertrophy is another hallmark of chronic cardiac allograft rejection. This pathologic hypertrophy is tightly linked to the immune cytokine IL-6, which promotes facets of chronic rejection in concert with TGF-beta and IL-17. These factors potentiate downstream mediators, such as CTGF, which promote the fibrosis associated with the disease. In this article, we summarize contemporary findings that have revealed several elements involved in the induction and progression of chronic rejection of cardiac allografts. Further efforts to elucidate the interplay between these factors may direct the development of targeted therapies for this disease.
Collapse
Affiliation(s)
- Adam J Booth
- Division of Pulmonary & Critical Care, Department of Internal Medicine, University of Michigan Medical Center, 6240 MSRBIII/0624, 1150 W Medical Center Drive, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
45
|
Chen M, Mohtize M, Matteï MF, Villemot JP, Kohler C, Faure GC, Béné MC, de Carvalho Bittencourt M. Reduced levels of both circulating CD4+ CD25+ CD127(low/neg) and CD4+ CD8(neg) invariant natural killer regulatory T cells in stable heart transplant recipients. Clin Exp Immunol 2010; 163:104-12. [PMID: 21039425 DOI: 10.1111/j.1365-2249.2010.04281.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A cross-regulation between two regulatory T cell (T(reg) ) subsets [CD4(+) CD25(+) and invariant natural killer (NK) T - iNK T] has been described to be important for allograft tolerance induction. However, few studies have evaluated these cellular subsets in stable recipients as correlates of favourable clinical outcome after heart transplantation. T(reg) and iNK T cell levels were assayed by flow cytometry in peripheral blood samples from 44 heart transplant recipients at a 2-year interval in 38 patients, and related to clinical outcome. Multi-parameter flow cytometry used CD4/CD25/CD127 labelling to best identify T(reg) , and a standard CD3/CD4/CD8/Vα24/Vβ11 labelling strategy to appreciate the proportions of iNK T cells. Both subtypes of potentially tolerogenic cells were found to be decreased in stable heart transplant recipients, with similar or further decreased levels after 2 years. Interestingly, the patient who presented with several rejection-suggesting incidents over this period displayed a greater than twofold increase of both cell subsets. These results suggest that CD4(+) CD25(+) CD127(low/neg) T(reg) and iNK T cells could be involved in the local control of organ rejection, by modulating immune responses in situ, in clinically stable patients. The measurement of these cell subsets in peripheral blood could be useful for non-invasive monitoring of heart transplant recipients, especially in the growing context of tolerance-induction trials.
Collapse
Affiliation(s)
- M Chen
- Laboratory of Immunology Department of Cardiothoracic Surgery, University Hospital of Nancy and Nancy Université, Allée du Morvan, Vandoeuvre-lès-Nancy, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Intermittent antibody-based combination therapy removes alloantibodies and achieves indefinite heart transplant survival in presensitized recipients. Transplantation 2010; 90:270-8. [PMID: 20571468 DOI: 10.1097/tp.0b013e3181e228bd] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND It is well established that primed/memory T cells play a critical role in heart transplant rejection. This contributes to the challenges faced in the transplant clinic because current treatments that are efficient in controlling naïve T cell alloresponses have limited efficacy on primed T cell responders. METHODS Fully MHC-mismatched heart transplantation was performed from BALB/c to C57BL/6 mice presensitized with BALB/c splenocytes 14 days pretransplantation. A combination therapy comprising CD70-, CD154-, and CD8-specific antibodies (Abs) was administered at day 0 and 4 posttransplantation with rapamycin on days 0 to 4. RESULTS The Ab combination therapy extended heart transplant survival in presensitized recipients from median survival time 8 days (MST) to MST 78 days. A decrease in the number of splenic interferon-gamma-secreting cells measured by ELISpot assay was seen in the treated group compared with the untreated controls. However, graft-infiltrating CD8+ and CD4+ T cells persisted despite treatment and the number of intragraft CD4+ T cells increased at day 30 posttransplantation. When an additional "rescue therapy" comprising the same Abs was readministered at days 30, 60, and 90 posttransplantation, T cell infiltration was reduced and indefinite graft survival was observed. Furthermore, rescue therapy resulted in gradual decrease in titer and, by day 90 posttransplantation, the complete loss of the preexisting, donor-specific Abs. CONCLUSION We conclude that our Ab combination therapy extends allograft survival in presensitized recipients. When combined with intermittent Ab-mediated rescue therapy, this results in indefinite allograft survival and a loss of the preexisting, donor-specific Abs from the circulation.
Collapse
|
47
|
Dinesh RK, Skaggs BJ, La Cava A, Hahn BH, Singh RP. CD8+ Tregs in lupus, autoimmunity, and beyond. Autoimmun Rev 2010; 9:560-8. [PMID: 20385256 DOI: 10.1016/j.autrev.2010.03.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 03/30/2010] [Indexed: 12/12/2022]
Abstract
While CD4(+)CD25(high) regulatory T cells (Tregs) have garnered much attention for their role in the maintenance of immune homeostasis, recent findings have shown that subsets of CD8(+) T cells (CD8(+) Tregs) display immunoregulatory functions as well. Both CD4(+) Tregs and CD8(+) Tregs appear impaired in number and/or function in several autoimmune diseases and in experimental animal models of autoimmunity, suggesting the possibility of immunotherapeutic targeting of these cells for improved management of autoimmune conditions. Our group has developed a strategy to induce CD8(+) Tregs in autoimmune mice through the use of a tolerogenic self-peptide, and new information has been gained on the phenotype, function and role of induced CD8(+) Tregs in autoimmunity. Here we present an overview of the role and mechanisms of action of CD8(+) Tregs in autoimmunity, with a special focus on lupus. We also discuss the potential role of CD8(+) Tregs in other diseases, including chronic infection and cancer.
Collapse
Affiliation(s)
- Ravi K Dinesh
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095-1670, USA
| | | | | | | | | |
Collapse
|
48
|
Zheng J, Liu Y, Lau YL, Tu W. CD40-activated B cells are more potent than immature dendritic cells to induce and expand CD4(+) regulatory T cells. Cell Mol Immunol 2010; 7:44-50. [PMID: 20081875 DOI: 10.1038/cmi.2009.103] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
CD4(+) regulatory T cells (Tregs) play an important role in maintaining immune tolerance by suppressing pathologic immune responses. The generation of large numbers of antigen-specific Tregs ex vivo is critical for the development of clinical immunotherapy based on the adoptive transfer of Tregs. Both CD40-activated B cells (CD40-B) and immature dendritic cells (imDCs) have been used as professional antigen-presenting cells (APCs) to generate antigen-specific Tregs. However, the efficiencies of CD40-B and imDCs to generate CD4(+) Tregs have not been compared directly and the mechanism driving the generation of these Tregs remains largely unknown. In this study, we found that CD40-B exhibited mature phenotypes and were more able to induce and expand CD4(high)CD25(+) Tregs than imDCs. Moreover, Tregs induced by CD40-B had greater suppressive capacity than those induced by imDCs. The generation of CD4(high)CD25(+) Tregs by CD40-B and imDCs is cell-cell contact dependent and partially relies on the expression of human leukocyte antigen (HLA)-DR and CD80/86. Differences in CD4(high)CD25(+) Treg generation efficiency were largely explained by the production of endogenous IL-2 by CD40-B. Our results suggest that CD40-B is better able to generate large numbers of antigen-specific Tregs than imDCs. Additionally, using CD40-B to generate Tregs may accelerate the clinical use of Treg-based immunotherapy in the treatment of allograft rejection, graft versus host disease (GVHD) and autoimmune diseases.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
49
|
Fuchs A, Atkinson JP, Fremeaux-Bacchi V, Kemper C. CD46-induced human Treg enhance B-cell responses. Eur J Immunol 2010; 39:3097-109. [PMID: 19784949 DOI: 10.1002/eji.200939392] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Regulatory CD4(+) T cells (Treg) are important modulators of the immune response. Different types of Treg have been identified based on whether they are thymically derived (natural Treg) or induced in the periphery (adaptive Treg). We recently reported on an adaptive Treg phenotype that can be induced by the concomitant stimulation of human CD4(+) T cells through CD3 and the membrane complement regulator CD46. These complement (CD46)-induced regulatory T cells (cTreg) potently inhibit bystander T-cell proliferation through high-level secretion of IL-10. In addition, cTreg express granzyme B and exhibit cytotoxic effects toward activated effector T cells. Here, we analyzed the effect of cTreg on B-cell functions in a co-culture system. We found that cTreg enhance B-cell Ab production. This B-cell support is dependent on cell/cell contact as well as cTreg-derived IL-10. In addition, we show that T cells from a CD46-deficient patient are not capable of promoting B-cell responses, whereas CD46-deficient B cells have no intrinsic defect in Ig production. This finding may relate to a subset of CD46-deficient patients, who present with common variable immunodeficiency. Thus, the lack of cTreg function in optimizing B-cell responses could explain why some CD46-deficient patients develop common variable immunodeficiency.
Collapse
Affiliation(s)
- Anja Fuchs
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | |
Collapse
|
50
|
Booth AJ, Csencsits-Smith K, Wood SC, Lu G, Lipson KE, Bishop DK. Connective tissue growth factor promotes fibrosis downstream of TGFbeta and IL-6 in chronic cardiac allograft rejection. Am J Transplant 2010; 10:220-30. [PMID: 19788504 PMCID: PMC2860022 DOI: 10.1111/j.1600-6143.2009.02826.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cardiac transplantation is an effective treatment for multiple types of heart failure refractive to therapy. Although immunosuppressive therapeutics have increased survival rates within the first year posttransplant, chronic rejection (CR) remains a significant barrier to long-term graft survival. Indicators of CR include patchy interstitial fibrosis, vascular occlusion and progressive loss of graft function. Multiple factors have been implicated in the onset and progression of CR, including TGFbeta, IL-6 and connective tissue growth factor (CTGF). While associated with CR, the role of CTGF in CR and the factors necessary for CTGF induction in vivo are not understood. To this end, we utilized forced expression and neutralizing antibody approaches. Transduction of allografts with CTGF significantly increased fibrotic tissue development, though not to levels observed with TGFbeta transduction. Further, intragraft CTGF expression was inhibited by IL-6 neutralization whereas TGFbeta expression remained unchanged, indicating that IL-6 effects may potentiate TGFbeta-mediated induction of CTGF. Finally, neutralizing CTGF significantly reduced graft fibrosis without reducing TGFbeta and IL-6 expression levels. These findings indicate that CTGF functions as a downstream mediator of fibrosis in CR, and that CTGF neutralization may ameliorate fibrosis and hypertrophy associated with CR.
Collapse
Affiliation(s)
- A. J. Booth
- Graduate Program in Immunology, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - K. Csencsits-Smith
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - S. C. Wood
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - G. Lu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | | | - D. K. Bishop
- Graduate Program in Immunology, University of Michigan Medical Center, Ann Arbor, MI, 48109, Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109,Corresponding Author: D. Keith Bishop, Ph. D. Transplant Immunology Research, Section of General Surgery, A560 MSRB II, Box 0654, University of Michigan Medical Center, 1150 W. Medical Center Dr. Ann Arbor, MI 48109, 734-763-0326 (Phone); 734-763-6199 (Fax),
| |
Collapse
|