1
|
Lee SK, Kwon JH, Jang JW, Bae SH, Yoon SK, Jung ES, Choi JY. The Critical Role of Regulatory T Cells in Immune Tolerance and Rejection Following Liver Transplantation: Interactions With the Gut Microbiome. Transplantation 2025; 109:784-793. [PMID: 39375899 DOI: 10.1097/tp.0000000000005220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Liver transplantation (LT) is the ultimate treatment for patients with end-stage liver disease or early hepatocellular carcinoma. In the context of LT, because of the unique immunological characteristics of human liver allograft, 5%-20% of selected LT recipients can achieve operational tolerance. Nonetheless, there remains a risk of rejection in LT patients. Maintaining immune homeostasis is thus crucial for improving clinical outcomes in these patients. In mechanism, several immune cells, including dendritic cells, Kupffer cells, myeloid-derived suppressor cells, hepatic stellate cells, regulatory B cells, and CD4 + regulatory T cells (Treg), contribute to achieving tolerance following LT. In terms of Treg, it plays a role in successfully minimizing immunosuppression or achieving tolerance post-LT while also reducing the risk of rejection. Furthermore, the gut microbiome modulates systemic immune functions along the gut-liver axis. Recent studies have explored changes in the microbiome and its metabolites under various conditions, including post-LT, acute rejection, and tolerance. Certain functional microbiomes and metabolites exhibit immunomodulatory functions, such as the augmentation of Treg, influencing immune homeostasis. Therefore, understanding the mechanisms of tolerance in LT, the role of Treg in tolerance and rejection, as well as their interactions with gut microbiome, is vital for the management of LT patients.
Collapse
Affiliation(s)
- Soon Kyu Lee
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Hyun Kwon
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sun Jung
- Department of Pathology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
2
|
van der List ACJ, Litjens NHR, Klepper M, Prevoo F, Betjes MGH. Progressive Loss of Donor-Reactive CD4 +Effector Memory T Cells due to Apoptosis Underlies Donor-Specific Hyporesponsiveness in Stable Renal Transplant Recipients. THE JOURNAL OF IMMUNOLOGY 2022; 209:1389-1400. [DOI: 10.4049/jimmunol.2200352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/01/2022] [Indexed: 11/06/2022]
|
3
|
Heterologous Immunity of Virus-Specific T Cells Leading to Alloreactivity: Possible Implications for Solid Organ Transplantation. Viruses 2021; 13:v13122359. [PMID: 34960628 PMCID: PMC8706157 DOI: 10.3390/v13122359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022] Open
Abstract
Exposure of the adaptive immune system to a pathogen can result in the activation and expansion of T cells capable of recognizing not only the specific antigen but also different unrelated antigens, a process which is commonly referred to as heterologous immunity. While such cross-reactivity is favourable in amplifying protective immune responses to pathogens, induction of T cell-mediated heterologous immune responses to allo-antigens in the setting of solid organ transplantation can potentially lead to allograft rejection. In this review, we provide an overview of murine and human studies investigating the incidence and functional properties of virus-specific memory T cells cross-reacting with allo-antigens and discuss their potential relevance in the context of solid organ transplantation.
Collapse
|
4
|
Samojlik MM, Stabler CL. Designing biomaterials for the modulation of allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes. Acta Biomater 2021; 133:87-101. [PMID: 34102338 PMCID: PMC9148663 DOI: 10.1016/j.actbio.2021.05.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
The effective suppression of adaptive immune responses is essential for the success of allogeneic cell therapies. In islet transplantation for Type 1 Diabetes, pre-existing autoimmunity provides an additional hurdle, as memory autoimmune T cells mediate both an autoantigen-specific attack on the donor beta cells and an alloantigen-specific attack on the donor graft cells. Immunosuppressive agents used for islet transplantation are generally successful in suppressing alloimmune responses, but dramatically hinder the widespread adoption of this therapeutic approach and fail to control memory T cell populations, which leaves the graft vulnerable to destruction. In this review, we highlight the capacity of biomaterials to provide local and nuanced instruction to suppress or alter immune pathways activated in response to an allogeneic islet transplant. Biomaterial immunoisolation is a common approach employed to block direct antigen recognition and downstream cell-mediated graft destruction; however, immunoisolation alone still permits shed donor antigens to escape into the host environment, resulting in indirect antigen recognition, immune cell activation, and the creation of a toxic graft site. Designing materials to decrease antigen escape, improve cell viability, and increase material compatibility are all approaches that can decrease the local release of antigen and danger signals into the implant microenvironment. Implant materials can be further enhanced through the local delivery of anti-inflammatory, suppressive, chemotactic, and/or tolerogenic agents, which serve to control both the innate and adaptive immune responses to the implant with a benefit of reduced systemic effects. Lessons learned from understanding how to manipulate allogeneic and autogenic immune responses to pancreatic islets can also be applied to other cell therapies to improve their efficacy and duration. STATEMENT OF SIGNIFICANCE: This review explores key immunologic concepts and critical pathways mediating graft rejection in Type 1 Diabetes, which can instruct the future purposeful design of immunomodulatory biomaterials for cell therapy. A summary of immunological pathways initiated following cellular implantation, as well as current systemic immunomodulatory agents used, is provided. We then outline the potential of biomaterials to modulate these responses. The capacity of polymeric encapsulation to block some powerful rejection pathways is covered. We also highlight the role of cellular health and biocompatibility in mitigating immune responses. Finally, we review the use of bioactive materials to proactively modulate local immune responses, focusing on key concepts of anti-inflammatory, suppressive, and tolerogenic agents.
Collapse
Affiliation(s)
- Magdalena M Samojlik
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Cherie L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA; Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
5
|
Karahan GE, Claas FHJ, Heidt S. Pre-existing Alloreactive T and B Cells and Their Possible Relevance for Pre-transplant Risk Estimation in Kidney Transplant Recipients. Front Med (Lausanne) 2020; 7:340. [PMID: 32793610 PMCID: PMC7385137 DOI: 10.3389/fmed.2020.00340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/08/2020] [Indexed: 12/25/2022] Open
Abstract
In allogeneic transplantation, genetic disparities between patient and donor may lead to cellular and humoral immune responses mediated by both naïve and memory alloreactive cells of the adaptive immune system. This review will focus on alloreactive T and B cells with emphasis on the memory compartment, their role in relation to kidney rejection, and in vitro assays to detect these alloreactive cells. Finally, the potential additional value of utilizing donor-specific memory T and B cell assays supplementary to current routine pre-transplant risk assessment of kidney transplant recipients will be discussed.
Collapse
Affiliation(s)
- Gonca E Karahan
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Sebastiaan Heidt
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
6
|
Ten Brinke A, Marek-Trzonkowska N, Mansilla MJ, Turksma AW, Piekarska K, Iwaszkiewicz-Grześ D, Passerini L, Locafaro G, Puñet-Ortiz J, van Ham SM, Hernandez-Fuentes MP, Martínez-Cáceres EM, Gregori S. Monitoring T-Cell Responses in Translational Studies: Optimization of Dye-Based Proliferation Assay for Evaluation of Antigen-Specific Responses. Front Immunol 2017; 8:1870. [PMID: 29312346 PMCID: PMC5742609 DOI: 10.3389/fimmu.2017.01870] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/08/2017] [Indexed: 01/05/2023] Open
Abstract
Adoptive therapy with regulatory T cells or tolerance-inducing antigen (Ag)-presenting cells is innovative and promising therapeutic approach to control undesired and harmful activation of the immune system, as observed in autoimmune diseases, solid organ and bone marrow transplantation. One of the critical issues to elucidate the mechanisms responsible for success or failure of these therapies and define the specificity of the therapy is the evaluation of the Ag-specific T-cell responses. Several efforts have been made to develop suitable and reproducible assays. Here, we focus on dye-based proliferation assays. We highlight with practical examples the fundamental issues to take into consideration for implementation of an effective and sensitive dye-based proliferation assay to monitor Ag-specific responses in patients. The most critical points were used to design a road map to set up and analyze the optimal assay to assess Ag-specific T-cell responses in patients undergoing different treatments. This is the first step to optimize monitoring of tolerance induction, allowing comparison of outcomes of different clinical studies. The road map can also be applied to other therapeutic interventions, not limited to tolerance induction therapies, in which Ag-specific T-cell responses are relevant such as vaccination approaches and cancer immunotherapy.
Collapse
Affiliation(s)
- Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Natalia Marek-Trzonkowska
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Maria J. Mansilla
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Annelies W. Turksma
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Karolina Piekarska
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Laura Passerini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Grazia Locafaro
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Joan Puñet-Ortiz
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - S. Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | | | - Eva M. Martínez-Cáceres
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
7
|
Fleischhauer K, Shaw BE. HLA-DP in unrelated hematopoietic cell transplantation revisited: challenges and opportunities. Blood 2017; 130:1089-1096. [PMID: 28667011 DOI: 10.1182/blood-2017-03-742346] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/17/2017] [Indexed: 11/20/2022] Open
Abstract
When considering HLA-matched hematopoietic cell transplantation (HCT), sibling and unrelated donors (UDs) are biologically different because UD-HCT is typically performed across HLA-DP disparities absent in sibling HCT. Mismatched HLA-DP is targeted by direct alloreactive T cell responses with important implications for graft-versus-host disease and graft-versus-leukemia. This concise review details special features of HLA-DP as model antigens for clinically permissive mismatches mediating limited T-cell alloreactivity with minimal toxicity, and describes future avenues for their exploitation in cellular immunotherapy of malignant blood disorders.
Collapse
Affiliation(s)
- Katharina Fleischhauer
- Institute for Experimental Cellular Therapy, Essen University Hospital, Essen, Germany
- German Cancer Consortium, Heidelberg, Germany; and
| | - Bronwen E Shaw
- Center for International Blood and Marrow Transplant Research, Froedtert & The Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
8
|
Stevens AM. Maternal microchimerism in health and disease. Best Pract Res Clin Obstet Gynaecol 2015; 31:121-30. [PMID: 26612343 DOI: 10.1016/j.bpobgyn.2015.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 08/31/2015] [Indexed: 12/19/2022]
Abstract
Circulating maternal cells transfer to the fetus during pregnancy, where they may integrate with the fetal immune and organ systems, creating a state of maternal microchimerism (MMc). MMc can persist throughout the child's life, and it has been implicated in the triggering or perpetuation of chronic inflammatory autoimmune diseases, in the context of specific major histocompatibility genes. Correlative data in humans have now been tested in animal model systems. Results suggest that maternal-fetal tolerance may have health implications far beyond the time of pregnancy and into the child's life.
Collapse
Affiliation(s)
- Anne M Stevens
- Department of Pediatrics, University of Washington, Seattle Children's Research Institute, 1900 9th Ave N, 9S-7, Seattle, WA 98101, USA.
| |
Collapse
|
9
|
NKG2D expression by CD8+ T cells contributes to GVHD and GVT effects in a murine model of allogeneic HSCT. Blood 2015; 125:3655-63. [PMID: 25788701 DOI: 10.1182/blood-2015-02-629006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/05/2015] [Indexed: 12/27/2022] Open
Abstract
In allogeneic hematopoietic stem cell transplantation (HSCT), controlling graft-versus-host disease (GVHD) while maintaining graft-versus-tumor (GVT) responses is of critical importance. Using a mouse model of allogeneic HSCT, we hereby demonstrate that NKG2D expression by CD8(+) T cells plays a major role in mediating GVHD and GVT effects by promoting the survival and cytotoxic function of CD8(+) T cells. The expression of NKG2D ligands was not induced persistently on normal tissues of allogeneic HSCT-recipient mice treated with anti-NKG2D antibody, suggesting that transient NKG2D blockade might be sufficient to attenuate GVHD and allow CD8(+) T cells to regain their GVT function. Indeed, short-term treatment with anti-NKG2D antibody restored GVT effects while maintaining an attenuated GVHD state. NKG2D expression was also detected on CD8(+) T cells from allogeneic HSCT patients and trended to be higher in those with active GVHD. Together, these data support a novel role for NKG2D expression by CD8(+) T cells during allogeneic HSCT, which could be potentially therapeutically exploited to separate GVHD from GVT effects.
Collapse
|
10
|
Nikoueinejad H, Sharif MR, Amirzargar A, Mirshafiey A, Einollahi B. Regulatory T Cells as a Therapeutic Tool To Induce Solid-Organ Transplant Tolerance: Current Clinical Experiences. EXP CLIN TRANSPLANT 2013; 11:379-87. [DOI: 10.6002/ect.2013.0004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Lynch R, Silva I, Chen B, Punch J, Cascalho M, Platt J. Cryptic B cell response to renal transplantation. Am J Transplant 2013; 13:1713-23. [PMID: 23750851 PMCID: PMC3764435 DOI: 10.1111/ajt.12308] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 03/13/2013] [Accepted: 03/17/2013] [Indexed: 01/25/2023]
Abstract
Transplantation reliably evokes allo-specific B cell and T cell responses in mice. Yet, human recipients of kidney transplants with normal function usually exhibit little or no antibody specific for the transplant donor during the early weeks and months after transplantation. Indeed, the absence of antidonor antibodies is taken to reflect effective immunosuppressive therapy and to predict a favorable outcome. Whether the absence of donor-specific antibodies reflects absence of a B cell response to the donor, tolerance to the donor or immunity masked by binding of donor-specific antibodies to the graft is not known. To distinguish between these possibilities, we devised a novel ELISPOT, using cultured donor, recipient and third-party fibroblasts as targets. We enumerated donor-specific antibody-secreting cells in the blood of nine renal allograft recipients with normal kidney function before and after transplantation. Although none of the nine subjects had detectable donor-specific antibodies before or after transplantation, all exhibited increases in the frequency of donor-specific antibody-secreting cells eight weeks after transplantation. The responses were directed against the donor HLA-class I antigens. The increase in frequency of donor-specific antibody-secreting cells after renal transplantation indicates that B cells respond specifically to the transplant donor more often than previously thought.
Collapse
Affiliation(s)
- R.J. Lynch
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - I.A. Silva
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - B.J. Chen
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - J.D. Punch
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - M. Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - J.L. Platt
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Yamada Y, Benichou G, Cosimi AB, Kawai T. Tolerance induction after organ transplantation, "delayed tolerance," via the mixed chimerism approach: planting flowers in a battle field. CHIMERISM 2013; 3:24-8. [PMID: 22690270 PMCID: PMC3370927 DOI: 10.4161/chim.20096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Yohei Yamada
- Massachusetts General Hospital, Transplant Center, Harvard Medical School, Boston, USA
| | | | | | | |
Collapse
|
13
|
Sagoo P, Lombardi G, Lechler RI. Relevance of regulatory T cell promotion of donor-specific tolerance in solid organ transplantation. Front Immunol 2012; 3:184. [PMID: 22811678 PMCID: PMC3395995 DOI: 10.3389/fimmu.2012.00184] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 06/14/2012] [Indexed: 01/29/2023] Open
Abstract
Current clinical strategies to control the alloimmune response after transplantation do not fully prevent induction of the immunological processes which lead to acute and chronic immune-mediated graft rejection, and as such the survival of a solid organ allograft is limited. Experimental research on naturally occurring CD4+CD25highFoxP3+ Regulatory T cells (Tregs) has indicated their potential to establish stable long-term graft acceptance, with the promise of providing a more effective therapy for transplant recipients. Current approaches for clinical use are based on the infusion of freshly isolated or ex vivo polyclonally expanded Tregs into graft recipients with an aim to redress the in vivo balance of T effector cells to Tregs. However mounting evidence suggests that regulation of donor-specific immunity may be central to achieving immunological tolerance. Therefore, the next stages in optimizing translation of Tregs to organ transplantation will be through the refinement and development of donor alloantigen-specific Treg therapy. The altering kinetics and intensity of alloantigen presentation pathways and alloimmune priming following transplantation may indeed influence the specificity of the Treg required and the timing or frequency at which it needs to be administered. Here we review and discuss the relevance of antigen-specific regulation of alloreactivity by Tregs in experimental and clinical studies of tolerance and explore the concept of delivering an optimal Treg for the induction and maintenance phases of achieving transplantation tolerance.
Collapse
Affiliation(s)
- Pervinder Sagoo
- Department Transplantation, Immunoregulation and Mucosal Biology, MRC Centre for Transplantation, King's College London London, UK
| | | | | |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW Here, we review the pathways of allorecognition and their potential relevance to the balance between regulatory and effector responses following transplantation. RECENT FINDINGS Transplantation between nonidentical members of the same species elicits an immune response that manifests as graft rejection or persistence. Presentation of foreign antigen to recipient T cells can occur via three nonmutually exclusive routes, the direct, indirect and semi-direct pathways. Allospecific T cells can have effector or regulatory functions, and the relative proportions of the two populations activated following alloantigen presentation are two of the factors that determine the clinical outcome. Regulatory T cells have been the subject of significant research, and there is now greater understanding of their recruitment and function in the context of allorecognition. SUMMARY A greater understanding of the mechanisms underlying allorecognition may be fundamental to appreciating how these different populations are recruited and could in turn inform novel strategies for immunomodulation.
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Here, we review the pathways of allorecognition and their potential relevance to the balance between regulatory and effector responses following transplantation. RECENT FINDINGS Transplantation between nonidentical members of the same species elicits an immune response that manifests as graft rejection or persistence. Presentation of foreign antigen to recipient T cells can occur via three nonmutually exclusive routes, the direct, indirect and semi-direct pathways. Allospecific T cells can have effector or regulatory functions, and the relative proportions of the two populations activated following alloantigen presentation are two of the factors that determine the clinical outcome. Regulatory T cells have been the subject of significant research, and there is now greater understanding of their recruitment and function in the context of allorecognition. SUMMARY A greater understanding of the mechanisms underlying allorecognition may be fundamental to appreciating how these different populations are recruited and could in turn inform novel strategies for immunomodulation.
Collapse
|
16
|
Yamada Y, Boskovic S, Aoyama A, Murakami T, Putheti P, Smith RN, Ochiai T, Nadazdin O, Koyama I, Boenisch O, Najafian N, Bhasin M, Colvin RB, Madsen JC, Strom TB, Sachs DH, Benichou G, Cosimi AB, Kawai T. Overcoming memory T-cell responses for induction of delayed tolerance in nonhuman primates. Am J Transplant 2012; 12:330-40. [PMID: 22053723 PMCID: PMC3268945 DOI: 10.1111/j.1600-6143.2011.03795.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The presence of alloreactive memory T cells is a major barrier for induction of tolerance in primates. In theory, delaying conditioning for tolerance induction until after organ transplantation could further decrease the efficacy of the regimen, since preexisting alloreactive memory T cells might be stimulated by the transplanted organ. Here, we show that such "delayed tolerance" can be induced in nonhuman primates through the mixed chimerism approach, if specific modifications to overcome/avoid donor-specific memory T-cell responses are provided. These modifications include adequate depletion of CD8+ memory T cells and timing of donor bone marrow administration to minimize levels of proinflammatory cytokines. Using this modified approach, mixed chimerism was induced successfully in 11 of 13 recipients of previously placed renal allografts and long-term survival without immunosuppression could be achieved in at least 6 of these 11 animals.
Collapse
Affiliation(s)
- Y. Yamada
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - S. Boskovic
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - A. Aoyama
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - T. Murakami
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - P. Putheti
- Department of Medicine, Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02114
| | - R. N. Smith
- Department of pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - T. Ochiai
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - O. Nadazdin
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - I. Koyama
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - O. Boenisch
- Department of Medicine, Renal Division, Brigham and Women’s Hospital, Boston, MA 02114
| | - N. Najafian
- Department of Medicine, Renal Division, Brigham and Women’s Hospital, Boston, MA 02114
| | - M.K. Bhasin
- Department of Medicine, Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02114
| | - R. B. Colvin
- Department of pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - J. C. Madsen
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - T. B. Strom
- Department of Medicine, Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02114
| | - D. H. Sachs
- Transplant Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - G. Benichou
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - A. B. Cosimi
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - T. Kawai
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
17
|
Fan H, Cao P, Game DS, Dazzi F, Liu Z, Jiang S. Regulatory T cell therapy for the induction of clinical organ transplantation tolerance. Semin Immunol 2011; 23:453-61. [PMID: 21920772 DOI: 10.1016/j.smim.2011.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The pursuit of transplantation tolerance is the holygrail in clinical organ transplantation. It has been established that regulatory T cells (Tregs) can confer donor-specific tolerance in mouse models of transplantation. However, this is crucially dependent on the strain combination, the organ transplanted and most importantly, the ratio of Tregs to alloreactive effector T cells. The ex vivo expansion of Tregs is one solution to increase the number of alloantigen specific cells capable of suppressing the alloresponse. Indeed, ex vivo expanded, alloantigen specific murine Tregs are shown to preferentially migrate to, and proliferate in, the graft and draining lymph node. In human transplantation it has been proposed that depletion of the majority of direct pathway alloreactive T cells will be required to tip the balance in favour of regulation. Ex vivo expansion of alloantigen specific, indirect pathway human Tregs, which can cross regulate the residual direct pathway has been established. Rapid expansion of these cells is possible, whilst they retain antigen specificity, suppressive properties and favourable homing markers. Furthermore, considerable progress has been made to define which immunosuppressive drugs favour the expansion and function of Tregs. Currently a series of clinical trials of adoptive Treg therapy in combination with depletion of alloreactive T cells and short term immunosuppression are underway for human transplantation with the aim of minimizing immunosuppressive drugs and completely withdrawal.
Collapse
Affiliation(s)
- Huimin Fan
- Shanghai East Hospital of Tongji University, Shanghai 200120, China
| | | | | | | | | | | |
Collapse
|
18
|
Cherkassky L, Lanning M, Lalli PN, Czerr J, Siegel H, Danziger-Isakov L, Srinivas T, Valujskikh A, Shoskes DA, Baldwin W, Fairchild RL, Poggio ED. Evaluation of alloreactivity in kidney transplant recipients treated with antithymocyte globulin versus IL-2 receptor blocker. Am J Transplant 2011; 11:1388-96. [PMID: 21564525 PMCID: PMC3226763 DOI: 10.1111/j.1600-6143.2011.03540.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Induction therapy is used in kidney transplantation to inhibit the activation of donor-reactive T cells which are detrimental to transplant outcomes. The choice of induction therapy is decided based on perceived immunological risk rather than by direct measurement of donor T-cell reactivity. We hypothesized that immune cellular alloreactivity pretransplantation can be quantified and that blocking versus depleting therapies have differential effects on the level of donor and third-party cellular alloreactivity. We studied 31 kidney transplant recipients treated with either antithymocyte globulin (ATG) or an IL-2 receptor blocker. We tested pre- and posttransplant peripheral blood cells by flow cytometry to characterize T-cell populations and by IFN-γ ELISPOT assays to assess the level of cellular alloreactivity. CD8(+) T cells were more resistant to depletion by ATG than CD4(+) T cells. Posttransplantation, frequencies of donor-reactive T cells were markedly decreased in the ATG-treated group but not in the IL-2 receptor blocker group, whereas the frequencies of third-party alloreactivity remained nearly equivalent. In conclusion, when ATG is used, marked and prolonged donor hyporesponsiveness with minimal effects on nondonor responses is observed. In contrast, induction with the IL-2 receptor blocker is less effective at diminishing donor T-cell reactivity.
Collapse
Affiliation(s)
- L. Cherkassky
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University
| | - M. Lanning
- Department of Immunology, Lerner Research Institute, Cleveland Clinic
| | - P. N. Lalli
- Allogen Laboratories, Inc,Transplant Center, Cleveland Clinic
| | - J. Czerr
- Department of Nephrology and Hypertension, Cleveland Clinic
| | - H. Siegel
- Department of Immunology, Lerner Research Institute, Cleveland Clinic
| | - L. Danziger-Isakov
- Department of Infectious Diseases, Cleveland Clinic,Transplant Center, Cleveland Clinic
| | - T. Srinivas
- Department of Nephrology and Hypertension, Cleveland Clinic,Transplant Center, Cleveland Clinic,Glickman Urological and Kidney Institute, Cleveland Clinic
| | - A. Valujskikh
- Department of Immunology, Lerner Research Institute, Cleveland Clinic,Transplant Center, Cleveland Clinic,Glickman Urological and Kidney Institute, Cleveland Clinic
| | - D. A. Shoskes
- Department of Urology, Cleveland Clinic,Transplant Center, Cleveland Clinic,Glickman Urological and Kidney Institute, Cleveland Clinic
| | - W. Baldwin
- Department of Immunology, Lerner Research Institute, Cleveland Clinic,Transplant Center, Cleveland Clinic
| | - R. L. Fairchild
- Department of Immunology, Lerner Research Institute, Cleveland Clinic,Transplant Center, Cleveland Clinic,Glickman Urological and Kidney Institute, Cleveland Clinic
| | - E. D. Poggio
- Department of Immunology, Lerner Research Institute, Cleveland Clinic,Department of Nephrology and Hypertension, Cleveland Clinic,Transplant Center, Cleveland Clinic,Glickman Urological and Kidney Institute, Cleveland Clinic
| |
Collapse
|
19
|
Abstract
Solid organ transplantation is the standard treatment to improve both the quality of life and survival in patients with various end-stage organ diseases. The primary barrier against successful transplantation is recipient alloimmunity and the need to be maintained on immunosuppressive therapies with associated side effects. Despite such treatments in renal transplantation, after death with a functioning graft, chronic allograft dysfunction (CAD) is the most common cause of late allograft loss. Recipient recognition of donor histocompatibility antigens, via direct, indirect, and semidirect pathways, is critically dependent on the antigen-presenting cell (APC) and elicits effector responses dominated by recipient T cells. In allograft rejection, the engagement of recipient and donor cells results in recruitment of T-helper (Th) cells of the Th1 and Th17 lineage to the graft. In cases in which the alloresponse is dominated by regulatory T cells (Tregs), rejection can be prevented and the allograft tolerated with minimum or no immunosuppression. Here, we review the pathways of allorecognition that underlie CAD and the T-cell effector phenotypes elicited as part of the alloresponse. Future therapies including depletion of donor-reactive lymphocytes, costimulation blockade, negative vaccination using dendritic cell subtypes, and Treg therapy are inferred from an understanding of these mechanisms of allograft rejection.
Collapse
|
20
|
Skelton TS, Kloc M, Ghobrial RM. Molecular and cellular pathways involved in the therapeutic functions of MHC molecules; a novel approach for mitigation of chronic rejection. ACTA ACUST UNITED AC 2011. [DOI: 10.4236/oji.2011.12003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
21
|
Tapirdamaz Ö, Mancham S, van der Laan LJW, Kazemier G, Thielemans K, Metselaar HJ, Kwekkeboom J. Detailed kinetics of the direct allo-response in human liver transplant recipients: new insights from an optimized assay. PLoS One 2010; 5:e14452. [PMID: 21206923 PMCID: PMC3012075 DOI: 10.1371/journal.pone.0014452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 11/18/2010] [Indexed: 12/17/2022] Open
Abstract
Conventional assays for quantification of allo-reactive T-cell precursor frequencies (PF) are relatively insensitive. We present a robust assay for quantification of PF of T-cells with direct donor-specificity, and establish the kinetics of circulating donor-specific T cells after liver transplantation (LTx). B cells from donor splenocytes were differentiated into professional antigen-presenting cells by CD40-engagement (CD40-B cells). CFSE-labelled PBMC from LTx-recipients obtained before and at several time points after LTx, were stimulated with donor-derived or 3rd party CD40-B cells. PF of donor-specific T cells were calculated from CFSE-dilution patterns, and intracellular IFN-γ was determined after re-stimulation with CD40-B cells. Compared to splenocytes, stimulations with CD40-B cells resulted in 3 to 5-fold higher responding T-cell PF. Memory and naïve T-cell subsets responded equally to allogeneic CD40-B cell stimulation. Donor-specific CD4+ and CD8+ T-cell PF ranged from 0.5 to 19% (median: 5.2%). One week after LTx, PF of circulating donor-specific CD4+ and CD8+ T cells increased significantly, while only a minor increase in numbers of T cells reacting to 3rd party allo-antigens was observed. One year after LTx numbers of CD4+ and CD8+ T cells reacting to donor antigens, as well as those reacting to 3rd party allo-antigens, were slightly lower compared to pre-transplant values. Moreover, CD4+ and CD8+ T cells responding to donor-derived, as well as those reacting to 3rd party CD40-B cells, produced less IFN-γ. In conclusion, our alternative approach enables detection of allo-reactive human T cells at high frequencies, and after application we conclude that donor-specific T-cell PF increase immediately after LTx. However, no evidence for a specific loss of circulating T-cells recognizing donor allo-antigens via the direct pathway up to 1 year after LTx was obtained, underscoring the relative insensitiveness of previous assays.
Collapse
Affiliation(s)
- Özlem Tapirdamaz
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Shanta Mancham
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Geert Kazemier
- Department of Surgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Herold J. Metselaar
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Quantitative and Functional Diversity of Cross-Reactive EBV-Specific CD8+ T Cells in a Longitudinal Study Cohort of Lung Transplant Recipients. Transplantation 2010; 90:1439-49. [DOI: 10.1097/tp.0b013e3181ff4ff3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
23
|
Golshayan D, Wyss JC, Buckland M, Hernandez-Fuentes M, Lechler RI. Differential role of naïve and memory CD4 T-cell subsets in primary alloresponses. Am J Transplant 2010; 10:1749-59. [PMID: 20659087 DOI: 10.1111/j.1600-6143.2010.03180.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The T cell response to major histocompatibility complex (MHC) alloantigens occurs via two main pathways. The direct pathway involves the recognition of intact allogeneic MHC:peptide complexes on donor cells and provokes uniquely high frequencies of responsive T cells. The indirect response results from alloantigens being processed like any other protein antigen and presented as peptide by autologous antigen-presenting cells. The frequencies of T cells with indirect allospecificity are orders of magnitude lower and comparable to other peptide-specific responses. In this study, we explored the contributions of naïve and memory CD4(+) T cells to these two pathways. Using an adoptive transfer and skin transplantation model we found that naive and memory CD4(+) T cells, both naturally occurring and induced by sensitization with multiple third-party alloantigens, contributed equally to graft rejection when only the direct pathway was operative. In contrast, the indirect response was predominantly mediated by the naïve subset. Elimination of regulatory CD4(+)CD25(+) T cells enabled memory cells to reject grafts through the indirect pathway, but at a much slower tempo than for naïve cells. These findings have implications for better targeting of immunosuppression to inhibit immediate and later forms of alloimmunity.
Collapse
Affiliation(s)
- D Golshayan
- Department of Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland.
| | | | | | | | | |
Collapse
|
24
|
Tanriver Y, Ratnasothy K, Bucy RP, Lombardi G, Lechler R. Targeting MHC class I monomers to dendritic cells inhibits the indirect pathway of allorecognition and the production of IgG alloantibodies leading to long-term allograft survival. THE JOURNAL OF IMMUNOLOGY 2010; 184:1757-64. [PMID: 20083658 DOI: 10.4049/jimmunol.0902987] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
T cell depletion strategies are an efficient therapy for the treatment of acute rejections and are an essential part of tolerance induction protocols in various animal models; however, they are usually nonselective and cause wholesale T cell depletion leaving the individual in a severely immunocompromised state. So far it has been difficult to selectively delete alloreactive T cells because the majority of protocols either delete all T cells, subsets of T cells, or subpopulations of T cells expressing certain activation markers, ignoring the Ag specificity of the TCR. We have developed a model in which we were able to selectively deplete alloreactive T cells with an indirect specificity by targeting intact MHC molecules to quiescent dendritic cells using 33D1 as the targeting Ab. This strategy enabled us to inhibit the indirect alloresponse against MHC-mismatched skin grafts and hence the generation of IgG alloantibodies, which depends on indirectly activated T cells. In combination with the temporary abrogation of the direct alloresponse, we were able to induce indefinite skin graft survival. Importantly, the targeting strategy had no detrimental effect on CD4(+)CD25(+)FoxP3(+) T cells, which could potentially be used as an adjunctive cellular therapy. Transplantation tolerance depends on the right balance between depletion and regulation. For the former this approach may be a useful tool in the development of future tolerance induction protocols in non-sensitized patients.
Collapse
Affiliation(s)
- Yakup Tanriver
- Medical Research Council Center for Transplantation, King's College London, School of Medicine, Guy's Hospital, London, UK
| | | | | | | | | |
Collapse
|
25
|
Macedo C, Orkis EA, Popescu I, Elinoff BD, Zeevi A, Shapiro R, Lakkis FG, Metes D. Contribution of naïve and memory T-cell populations to the human alloimmune response. Am J Transplant 2009; 9:2057-66. [PMID: 19624567 DOI: 10.1111/j.1600-6143.2009.02742.x] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
T-cell alloimmunity plays a dominant role in allograft rejection. The precise contribution of naïve and memory T cells to this response however remains unclear. To address this question, we established an ex vivo flow-cytometric assay that simultaneously measures proliferation, precursor frequency and effector molecule (IFNgamma, granzyme B/perforin) production of alloreactive T cells. By applying this assay to peripheral blood mononuclear cells from healthy volunteers, we demonstrate that the CD4+ and CD8+ populations mount similar proliferative responses and contain comparable frequencies of alloreactive precursors. Effector molecule expression, however, was significantly higher among CD8+ T cells. Analysis of sorted naïve and memory T cells showed that alloreactive precursors were equally present in both populations. The CD8+ effector and terminally differentiated effector memory subsets contained the highest proportion of granzyme B/perforin after allostimulation, suggesting that these cells present a significant threat to transplanted organs. Finally, we demonstrate that virus-specific lymphocytes contribute significantly to the alloresponse in certain responder-stimulator HLA combinations, underscoring the importance of T-cell cross-reactivity in alloimmunity. These results provide a quantitative assessment of the roles of naïve and memory T-cell subsets in the normal human alloimmune response and establish a platform for measuring T-cell alloreactivity pre- and posttransplantation.
Collapse
Affiliation(s)
- C Macedo
- Human Immunology Program, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Bestard O, Nickel P, Cruzado JM, Schoenemann C, Boenisch O, Sefrin A, Grinyó JM, Volk HD, Reinke P. Circulating alloreactive T cells correlate with graft function in longstanding renal transplant recipients. J Am Soc Nephrol 2008; 19:1419-29. [PMID: 18417724 DOI: 10.1681/asn.2007050539] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Monitoring for alloreactive memory T cells after organ transplantation may allow individualization of immunosuppression. Two pathways of T cell allorecognition have been implicated in chronic graft dysfunction: Direct (recipient T cells respond to donor peptides presented by donor antigen-presenting cells) and indirect (donor peptides are processed and presented by recipient antigen-presenting cells). Previous studies have assessed these alloresponses only during the first 2 yr after kidney transplantation,so this study correlated the presence of circulating donor-reactive memory/effector T cells, primed by both pathways, in 34 longstanding living-donor renal transplant recipients using the highly sensitive IFN-gamma Elispot assay. Remarkably, 59% of patients had directly primed donor-reactive T cells, and their presence correlated directly with serum creatinine (P = 0.001) and inversely with estimated GFR (P = 0.042). Multivariate analysis revealed that hyporesponsiveness of direct, donor-specific T cells was the only variable that significantly correlated with graft function and that antidonor indirect alloreactivity was the only variable that significantly correlated with proteinuria. Interestingly, when both allorecognition pathways were considered together, patients with undetectable direct alloreactivity had better longterm graft function, independent of allosensitization by the indirect pathway. In conclusion, circulating donor-specific alloreactive T cells primed by both pathways are detectable long after transplantation and are associated with graft injury. Assessment of alloreactive memory/effector T cells might be helpful to tailor individual immunosuppression regimens for transplant recipients in the future.
Collapse
Affiliation(s)
- Oriol Bestard
- Nephrology Department, Hospital Universitari de Bellvitge, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Afzali B, Lechler RI, Hernandez-Fuentes MP. Allorecognition and the alloresponse: clinical implications. ACTA ACUST UNITED AC 2007; 69:545-56. [PMID: 17498264 DOI: 10.1111/j.1399-0039.2007.00834.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The artificial transfer of tissues or cells between genetically diverse individuals elicits an immune response that is adaptive and specific. This response is orchestrated by T lymphocytes that are recognizing, amongst others, major histocompatibility complex (MHC) molecules expressed on the surface of the transferred cells. Three pathways of recognition are described: direct, indirect and semi-direct. The sets of antigens that are recognized in this setting are also discussed, namely, MHC protein products, the MHC class I-related chain (MIC) system, minor histocompatibility antigens and natural killer cell receptor ligands. The end product of the effector responses are hyperacute, acute and chronic rejection. Special circumstances surround the situation of pregnancy and bone marrow transplantation because in the latter, the transferred cells are the ones originating the immune response, not the host. As the understanding of these processes improves, the ability to generate clinically viable immunotherapies will increase.
Collapse
Affiliation(s)
- B Afzali
- Department of Nephrology and Transplantation, King's College London, Guy's Hospital Campus, London, UK
| | | | | |
Collapse
|
28
|
Stevens AM. Do maternal cells trigger or perpetuate autoimmune diseases in children? Pediatr Rheumatol Online J 2007; 5:9. [PMID: 17550578 PMCID: PMC1892552 DOI: 10.1186/1546-0096-5-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Accepted: 05/16/2007] [Indexed: 02/04/2023] Open
Abstract
The placental barrier is not the impenetrable wall that it was once presumed to be. During pregnancy, fetal cells pass into the mother, where they persist for decades after the pregnancy, leading to fetal microchimerism (FMc). Maternal cells also pass into the fetus, where they can persist long after birth of the child into adulthood, leading to maternal microchimerism(MMc). FMc and MMc represent foreign cells, and thus have been implicated in the pathogenesis of autoimmune diseases that resemble graft-versus-host disease after stem cell transplantation. FMc, hypothesized to contribute to the high predisposition of autoimmune diseases in women, has been reviewed recently. In patients who have never been pregnant, (children, males, and nulliparous females), MMc may represent the foreign cells that initiate or perpetuate chronic inflammatory disease.
Collapse
Affiliation(s)
- Anne M Stevens
- Department of Pediatrics, University of Washington, Children's Hospital and Regional Medical Center, 307 Westlake Ave N, Suite 300, Seattle, WA 98109, USA.
| |
Collapse
|
29
|
Ferraris JR, Tambutti M, Prigoshin N. Improved long-term graft function in kidney transplant recipients with donor antigen-specific hyporeactivity. Pediatr Transplant 2007; 11:139-44. [PMID: 17300492 DOI: 10.1111/j.1399-3046.2006.00621.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We investigated the development of donor antigen-specific hyporeactivity by using donor cells as stimulator cells in the MLC and comparing the pre- and post-transplant responses of peripheral blood mononuclear cells. Twenty-two haploidentical pediatric living-relative donor recipients treated with daclizumab, methylprednisone, mofetil mycophenolate and calcineurin inhibitors were tested for study. Of these, 50% of the recipients developed in vitro donor antigen-specific hyporeactivity. The recipients who did so have higher creatinine clearance levels at 12, 24 and 36 months post-transplant (104, 92 and 81 mL/min/1.73 m(2), respectively) than those who remained responsive to donor antigens (77, 74 and 70 mL/min/1.73 m(2)) (p < 0.05). Acute rejection episodes were not observed; however, no recipients with donor-specific hyporeactivity have been diagnosed with CAN, unlike three recipients who remained responsive to donor antigens (0% vs. 27.3%, p = 0.06). Differences in accumulative doses of methylprednisone and mofetil mycophenolate were observed between hyporeactivity- and response-patients to donor antigens at the three years end-point (1.9 +/- 0.8 g/m(2) vs. 4.2 +/- 0.5 g/m(2), and 277 +/- 89 g/m(2) vs. 672 +/- 16.0 g/m(2); p < 0.01 and <0.02, respectively). We conclude that the development of donor antigen-specific hyporeactivity correlate with improved graft function and may permit lower immunosuppression.
Collapse
Affiliation(s)
- Jorge R Ferraris
- Sección Trasplante Renal Pediátrico y Servicio de Histocompatibilidad, Buenos Aires, Argentina.
| | | | | |
Collapse
|
30
|
Golshayan D, Buhler L, Lechler RI, Pascual M. From current immunosuppressive strategies to clinical tolerance of allografts. Transpl Int 2007; 20:12-24. [PMID: 17181648 DOI: 10.1111/j.1432-2277.2006.00401.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In order to prevent allograft rejection, most current immunosuppressive drugs nonspecifically target T-cell activation, clonal expansion or differentiation into effector cells. Experimental models have shown that it is possible to exploit the central and peripheral mechanisms that normally maintain immune homeostasis and tolerance to self-antigens, in order to induce tolerance to alloantigens. Central tolerance results from intrathymic deletion of T cells with high avidity for thymically expressed antigens. Peripheral tolerance to nonself-molecules can be achieved by various mechanisms including deletion of activated/effector T cells, anergy induction and active regulation of effector T cells. In this article, we briefly discuss the pathways of allorecognition and their relevance to current immunosuppressive strategies and to the induction of transplantation tolerance (through haematopoietic mixed chimerism, depleting protocols, costimulatory blockade and regulatory T cells). We then review the prospect of clinical applicability of these protocols in solid organ transplantation.
Collapse
Affiliation(s)
- Dela Golshayan
- Transplantation Centre and Transplantation Immunopathology Laboratory, University Hospital Lausanne, Lausanne, Switzerland.
| | | | | | | |
Collapse
|
31
|
Korin YD, Lee C, Gjertson DW, Wilkinson AH, Pham TP, Danovitch GM, Gritsch HA, Reed EF. A novel flow assay for the detection of cytokine secreting alloreactive T cells: application to immune monitoring. Hum Immunol 2006; 66:1110-24. [PMID: 16571412 DOI: 10.1016/j.humimm.2005.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2005] [Accepted: 10/13/2005] [Indexed: 11/26/2022]
Abstract
The direct and indirect allorecognition pathways play an important role in graft rejection. We hypothesized that the presence of alloreactive memory T cells in the recipient's circulation increases the risk of rejection after transplantation. The objective of this study was to develop a noninvasive, immune monitoring tool that simultaneously measures donor-specific responses via both the direct and indirect recognition pathways. Our laboratory developed a whole blood flow cytometric cytokine secretion assay to identify interferon (IFN)-gamma secreting memory T cells in whole blood of renal transplant patients. The assay readily detected IFN-gamma producing CD3+ T cells in response to recall antigens tetanus toxoid, purified protein derivative, and alloantigens in whole blood from healthy controls. Analysis of sequential posttransplant blood samples from 19 renal allograft recipients showed that alloimmune responses were higher in transplant recipients who had undergone acute rejection than in those without acute rejection episodes. In addition, patients showing increased creatinine levels 3 months after transplantation were more likely to exhibit alloimmune responses than recipients with stable graft function. The flow cytokine secretion assay provides a reliable and simple method for identification of patients at risk of acute rejection and early graft dysfunction.
Collapse
Affiliation(s)
- Yael D Korin
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Herrera OB, Golshayan D, Tibbott R, Salcido Ochoa F, James MJ, Marelli-Berg FM, Lechler RI. A novel pathway of alloantigen presentation by dendritic cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:4828-37. [PMID: 15470023 DOI: 10.4049/jimmunol.173.8.4828] [Citation(s) in RCA: 234] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In the context of transplantation, dendritic cells (DCs) can sensitize alloreactive T cells via two pathways. The direct pathway is initiated by donor DCs presenting intact donor MHC molecules. The indirect pathway results from recipient DCs processing and presenting donor MHC as peptide. This simple dichotomy suggests that T cells with direct and indirect allospecificity cannot cross-regulate each other because distinct APCs are involved. In this study we describe a third, semidirect pathway of MHC alloantigen presentation by DCs that challenges this conclusion. Mouse DCs, when cocultured with allogeneic DCs or endothelial cells, acquired substantial levels of class I and class II MHC:peptide complexes in a temperature- and energy-dependent manner. Most importantly, DCs acquired allogeneic MHC in vivo upon migration to regional lymph nodes. The acquired MHC molecules were detected by Ab staining and induced proliferation of Ag-specific T cells in vitro. These data suggest that recipient DCs, due to acquisition of donor MHC molecules, may link T cells with direct and indirect allospecificity.
Collapse
|
33
|
|
34
|
Game DS, Hernandez-Fuentes MP, Chaudhry AN, Lechler RI. CD4+CD25+ regulatory T cells do not significantly contribute to direct pathway hyporesponsiveness in stable renal transplant patients. J Am Soc Nephrol 2003; 14:1652-61. [PMID: 12761268 DOI: 10.1097/01.asn.0000067411.03024.a9] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
CD4(+)CD25(+) regulatory T cells have been shown to regulate a variety of autoimmune and allogeneic responses in mice and humans. The role of CD4(+)CD25(+) cells in regulating alloresponses in human transplant recipients remains uncertain. Previous research has demonstrated a reduced frequency of direct pathway donor-specific T cells in renal transplant recipients when compared with the frequency of T cells reactive to an HLA-matched third party. A number of mechanisms have been proposed to account for this finding; the purpose of this study was to determine whether CD4(+)CD25(+) cells play a significant role. Twelve stable renal transplant patients were investigated using limiting dilution assay (LDA) and ELISPOT for interferon-gamma to determine the effect of depleting CD4(+)CD25(+) cells on the direct pathway alloresponse. The percentage of CD4(+)CD25(+) cells in the peripheral blood of the study patients was equivalent to that of healthy controls. Furthermore, in no case did depletion of CD4(+)CD25(+) cells significantly increase the frequency of donor-specific T cells detected by LDA. This was also found with ELISPOT in all except one patient, in whom depletion revealed an increased frequency of alloreactive T cell to both donor and third party. Finally, kinetic analysis of the LDA data did not indicate regulation against donor when compared with third party. It is concluded that the action of CD4(+)CD25(+) regulatory cells is not the main mechanism of donor-specific hyporesponsiveness in the direct pathway of allorecognition.
Collapse
Affiliation(s)
- David S Game
- Department of Immunology, Imperial College Faculty of Medicine, Hammersmith Hospital, London, UK
| | | | | | | |
Collapse
|
35
|
Abstract
Transplant biologists have made significant progress over the last 20 years towards unraveling the immunologic intricacies of allograft rejection. This large body of work has resulted in an improved understanding of T-cell allorecognition at a molecular level and has provided new insight into the functional consequences resulting from the allorecognition events. The findings suggest that the survival and the histologic features of a transplanted organ are influenced not only by the T-cell recognition pathway, but also by the frequency, the induced effector functions and the specific cellular targets of the alloreactive T-cell repertoire.
Collapse
Affiliation(s)
- Peter S Heeger
- Department of Immunology and The Glickman Urologic Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
36
|
Sijpkens YWJ, Doxiadis IIN, Mallat MJK, de Fijter JW, Bruijn JA, Claas FHJ, Paul LC. Early versus late acute rejection episodes in renal transplantation. Transplantation 2003; 75:204-8. [PMID: 12548124 DOI: 10.1097/01.tp.0000041722.34000.21] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Acute rejection is a major complication after renal transplantation and the most important risk factor for chronic rejection. We investigated whether the timing of the last treated acute rejection episode (ARE) influences long-term outcome and compared the risk profiles of early versus late ARE. METHODS A cohort of 654 patients who underwent cadaveric renal transplants (1983-1997) that functioned for more than 6 months was studied. In 384 of 654 transplant recipients, one or more treated AREs were documented; the last ARE occurred in 297 of 384 transplant recipients within 3 months and in 87 of 384 after 3 months. Applying multivariate logistic regression analysis, we compared the predictor variables of the two groups with transplants without AREs. RESULTS Ten-year graft survival rates censored for causes of graft loss other than chronic rejection were 94%, 86%, and 45% for patients without ARE, with early ARE, and with late ARE, respectively. Delayed graft function, odds ratio (OR) 2.37 (1.55-3.62), and major histocompatibility complex (MHC) class II incompatibility, OR 2.28 (1.62-3.20) per human leukocyte antigen (HLA)-DR mismatch, were independent risk factors for early ARE. In contrast, recipient age, OR 0.75 (0.61-0.93) per 10-year increase, donor age, OR 1.28 (1.07-1.53) per 10-year increase, female donor gender, OR 1.74 (1.03-2.94), and MHC class I incompatibility, OR 1.35 (1.07-1.72) per mismatch of cross reactive groups, were associated with late ARE. CONCLUSIONS Late ARE has a detrimental impact on long-term graft survival and is associated with MHC class I incompatibility, whereas early ARE is correlated with HLA-DR mismatches and has a better prognosis. These data are consistent with the role of direct and indirect allorecognition in the pathophysiology of early and late ARE, respectively.
Collapse
Affiliation(s)
- Yvo W J Sijpkens
- Department of Nephrology, Leiden University Medical Center, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
37
|
Carreno MR, Fuller L, Mathew JM, Ciancio G, Burke GW, Esquenazi V, Ricordi C, Tzakis AG, Miller J. Human donor bone marrow cells induce in vitro "suppressor T cells" that functionally suppress autologous B cells. Hum Immunol 2003; 64:21-30. [PMID: 12507811 DOI: 10.1016/s0198-8859(02)00774-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have reported a beneficial effect of donor vertebral body bone marrow cells (DBMC) infusions in cadaver renal allograft recipients in a 6-year follow-up, but with a transient increase in early (6 month) postoperative CMV infections and concomitant suppressed immunoglobulins (Ig) production. We also found that although there was no difference between the DBMC-infused and non-infused (control) groups in the development of donor-specific antibody, we now describe an additional difference seen in the percent reactive antibody (PRA) reactivity against a panel of HLA antigens that developed postoperatively. We hypothesize that (allogeneic) antigen presenting cells in the DBMC, systemically infused, caused the generation of recipient T suppressor (T4-suppressor) cells, thereby "inducing" a negative influence on B cell Ig production. We tested this notion in vitro by incubating PBL from CMV IgG positive laboratory volunteers with either (allogeneic) T-cell depleted DBMC or donor spleen cells (DSPC) from (the same) cadaver donors. After 7 days, the (responding) T cells were collected using magnetic beads and placed in culture with purified B cells freshly obtained from the same (autologous) CMV positive volunteer. To these cultures were added either media or 40 ng of CMV antigen. After 3, 5, 7, and 9 days, the expression of surface anti-CMV Ig was measured by flow cytometry using a panel of fluorescent markers double-labeled for activated B cells (CD20, CD19, and HLA DRw) and CMV-FITC. We also determined the phenotype of the cultured T cells using anti-CD3, CD4, and CD62L specific monoclonal antibodies. B cells that had been in contact with autologous T cells derived from DBMC cultures (TBM) were less likely to express anti-CMV surface Ig than those cultured with DSPC (TSP). The flow cytometry analysis revealed an increase in the number of T4 suppressor cells (CD3+, CD4+, CD62L+) in the TBM group, whereas the T4 helper phenotype (CD3+, CD4+, CD62L-) predominated in the TSP group. These in vitro findings support the notion that (allogeneic) DBMC infusions can induce a T4 suppressor (regulatory) influence and thereby indirectly affect B-cell function.
Collapse
Affiliation(s)
- Manuel R Carreno
- Departments of Surgery, Division of Transplantation, University of Miami School of Medicine, Miami, FL 33101, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Masuyama JI, Kaga S, Kano S, Minota S. A novel costimulation pathway via the 4C8 antigen for the induction of CD4+ regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2002; 169:3710-6. [PMID: 12244164 DOI: 10.4049/jimmunol.169.7.3710] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD4(+)CD25(+) regulatory T (Treg) cells naturally occur in mice and humans, and similar Treg cells can be induced in vivo and in vitro. However, the molecular mechanisms that mediate the generation of these Treg cell populations remain unknown. We previously described anti-4C8 mAbs that inhibit the postadhesive transendothelial migration of T cells through human endothelial cell monolayers. We demonstrate in this work that Treg cells are induced by costimulation of CD4(+) T cells with anti-CD3 plus anti-4C8. The costimulation induced full activation of CD4(+) T cells with high levels of IL-2 production and cellular expansion that were comparable to those obtained on costimulation by CD28. However, upon restimulation, 4C8-costimulated cells produced high levels of IL-10 but no IL-2 or IL-4, and maintained high expression levels of CD25 and intracellular CD152, as compared to CD28-costimulated cells. The former cells showed hyporesponsiveness to anti-CD3 stimulation and suppressed the activation of bystander T cells depending on cell contact but not IL-10 or TGF-beta. The suppressor cells developed from CD4(+)CD25(-)CD45RO(+) cells. The results suggest that 4C8 costimulation induces the generation of Treg cells that share phenotypic and functional features with CD4(+)CD25(+) T cells, and that CD25(-) memory T cells may differentiate into certain Treg cell subsets in the periphery.
Collapse
Affiliation(s)
- Jun-Ichi Masuyama
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Jichi Medical School, Tochigi, Japan.
| | | | | | | |
Collapse
|
39
|
Gebauer BS, Hricik DE, Atallah A, Bryan K, Riley J, Tary-Lehmann M, Greenspan NS, Dejelo C, Boehm BO, Hering BJ, Heeger PS. Evolution of the enzyme-linked immunosorbent spot assay for post-transplant alloreactivity as a potentially useful immune monitoring tool. Am J Transplant 2002; 2:857-66. [PMID: 12392292 DOI: 10.1034/j.1600-6143.2002.20908.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Post-transplant monitoring of cellular immunity has the potential to guide alterations in medical therapy. To this end, our laboratory has developed an enzyme-linked immunosorbent spot (ELISPOT) assay for detection of peripheral blood alloimmunity. Peripheral blood lymphocytes (PBLs) from normal volunteers and from renal allograft recipients were tested against donor stimulator cells for their ability to respond in 'one-way' cytokine ELISPOT assays. T cell depletion of donor spleen or PBLs eliminated donor cell cytokine secretion while preserving the ability of these cells to present allo-antigen to responding T cells. Alloreactive IFN-gamma-producing PBLs derive from the memory T cell pool and are readily detectable in recipients of renal allografts taking immunosuppressant medications. A significant expansion of IFN-gamma-producing donor-reactive memory PBLs was detectable at 4-6 months post-transplant in those who had experienced an acute rejection episode compared with those with a stable post-transplant course. The data demonstrate the feasibility of repeated post-transplant monitoring of allograft recipients, and provide the foundation for improving the care of human transplant recipients through rational clinical decision-making based on measures of immune function.
Collapse
Affiliation(s)
- Britta S Gebauer
- Department of Surgery, University of Minnesota School of Medicine, Minneapolis, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Allorecognition occurs when the host immune system detects same-species, non-self antigens and this is the trigger for allograft rejection. Host T cells detect these 'foreign' antigens which are mostly derived from a highly polymorphic region of the genome called the major histocompatibility complex. Allorecognition can occur by two distinct, but not mutually exclusive pathways: direct and indirect. The direct pathway results from the recognition of foreign major histocompatibility molecules, intact, on the surface of donor cells. Indirect allorecognition occurs when donor histocompatibility molecules are internalised, processed, and presented as peptides by host antigen presenting cells--this is the manner in which the immune system normally sees antigen. However, in addition to antigen recognition, T cell activation requires the provision of costimulatory signals, the prerogative of bone marrow-derived, specialised antigen-presenting cells (APC). Once these have been depleted from a transplanted organ, as occurs within weeks of transplantation, the parenchymal cells of the transplant are incapable of driving direct pathway activation of recipient T cells. Alloantigen recognition on these non-professional APCs may have a tolerising effect and indeed, the frequency of T cells reactive to the direct pathway diminishes with time irrespective of whether or not chronic transplant rejection occurs. This implies that while the direct pathway plays a dominant role in acute rejection, it is unlikely to contribute to chronic rejection. Assays of T cell responses have, however, found an association between the indirect pathway and chronic rejection and animal models support a role for the indirect pathway in both acute and chronic rejection. The indirect pathway is likely to be permanently active due to traffic of recipient APCs through the graft. The challenge that this poses in the pursuit of clinical tolerance is how to induce tolerance in T cells with indirect allospecificity. The answer may lie in manipulation of the environment of the interaction between the T cell and APC. Apart from recognition without costimulation, there are other circumstances when recognition without activation can occur although the in vivo relevance is uncertain. The presence of regulatory cytokines or inhibitory surface molecules either from a distinct regulatory cell, or as a negative feedback loop may prevent activation; this could also happen without sufficient stimulatory support: the final outcome is likely to be decided by the overall balance. Furthermore, some peptides may act as antagonists to T cell activation, usually when the agonist peptide is structurally very similar. It is hoped that the careful study of these mechanisms will reveal ways of ensuring allorecognition without activation and thus donor-specific tolerance.
Collapse
Affiliation(s)
- David S Game
- Department of Immunology, Imperial College School of Medicine, London, UK
| | | |
Collapse
|
41
|
|
42
|
Baker RJ, Hernandez-Fuentes MP, Brookes PA, Chaudhry AN, Cook HT, Lechler RI. Loss of direct and maintenance of indirect alloresponses in renal allograft recipients: implications for the pathogenesis of chronic allograft nephropathy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:7199-206. [PMID: 11739543 DOI: 10.4049/jimmunol.167.12.7199] [Citation(s) in RCA: 170] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chronic allograft nephropathy (CAN) is the principal cause of late renal allograft failure. This complex process is multifactorial in origin, and there is good evidence for immune-mediated effects. The immune contribution to this process is directed by CD4(+) T cells, which can be activated by either direct or indirect pathways of allorecognition. For the first time, these pathways have been simultaneously compared in a cohort of 22 longstanding renal allograft recipients (13 with good function and nine with CAN). CD4(+) T cells from all patients reveal donor-specific hyporesponsiveness by the direct pathway according to proliferation or the secretion of the cytokines IL-2, IL-5, and IFN-gamma. Donor-specific cytotoxic T cell responses were also attenuated. In contrast, the frequencies of indirectly alloreactive cells were maintained, patients with CAN having significantly higher frequencies of CD4(+) T cells indirectly activated by allogeneic peptides when compared with controls with good allograft function. An extensive search for alloantibodies has revealed significant titers in only a minority of patients, both with and without CAN. In summary, this study demonstrates widespread donor-specific hyporesponsiveness in directly activated CD4(+) T cells derived from longstanding recipients of renal allografts, whether they have CAN or not. However, patients with CAN have significantly higher frequencies of CD4(+) T cells activated by donor Ags in an indirect manner, a phenomenon resembling split tolerance. These findings provide an insight into the pathogenesis of CAN and also have implications for the development of a clinical tolerance assay.
Collapse
Affiliation(s)
- R J Baker
- Department of Immunology, Faculty of Medicine, Imperial College, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
43
|
Lechler R, Chai JG, Marelli-Berg F, Lombardi G. The contributions of T-cell anergy to peripheral T-cell tolerance. Immunology 2001; 103:262-9. [PMID: 11454055 PMCID: PMC1783252 DOI: 10.1046/j.1365-2567.2001.01250.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- R Lechler
- Department of Immunology, Imperial College School of Medicine, London, UK.
| | | | | | | |
Collapse
|