1
|
Zhanzak Z, Johnson AC, Foster P, Cardenas MA, Morris AB, Zhang J, Karadkhele G, Badell IR, Morris AA, Au-Yeung BB, Roversi FM, Silva JAF, Breeden C, Hadley A, Zhang W, Larsen CP, Kissick HT. Identification of indirect CD4 + T cell epitopes associated with transplant rejection provides a target for donor-specific tolerance induction. Immunity 2025; 58:448-464.e6. [PMID: 39889703 DOI: 10.1016/j.immuni.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/24/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
Antibodies against the donor human leukocyte antigen (HLA) molecules drive late transplant failure, with HLA-DQ donor-specific antibodies (DSAs) posing the highest rejection risk. Here, we investigated the role of indirect CD4+ T cell epitopes-donor-derived peptides presented by recipient major histocompatibility complex (MHC) class II-in DSA formation. Antigen mapping of samples from HLA-DQ DSA-positive kidney and heart transplant recipients revealed two polymorphic hotspots in donor HLA-DQ that generated alloreactive peptides. Antigen mapping of indirect CD4+ T cell epitopes in a mouse model of fully MHC mismatched skin graft transplantation (BALB/c to C57BL/6) identified a similar epitope (amino acids 287-301) derived from the donor H2-Kd. Tetramer-binding Kd287+ CD4+ T cells were detected during rejection and their transfer into T cell-deficient mice induced DSA. Systemic delivery of high-dose donor H2-Kd peptides combined with CTLA4-Ig reduced the frequencies of Kd287+ CD4+ T cells and DSA formation. Thus, targeting a narrow range of donor antigens may prevent DSA formation and improve transplant outcomes.
Collapse
Affiliation(s)
- Zhuldyz Zhanzak
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Aileen C Johnson
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Petra Foster
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Maria A Cardenas
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Anna B Morris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Joan Zhang
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Geeta Karadkhele
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - I Raul Badell
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Alanna A Morris
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Byron B Au-Yeung
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Division of Immunology, Department of Medicine, Lowance Center for Human Immunology, Emory University School of Medicine Atlanta, GA, USA
| | - Fernanda M Roversi
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Juliete A F Silva
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Cynthia Breeden
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Annette Hadley
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Weiwen Zhang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Christian P Larsen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA; Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Haydn T Kissick
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA; Winship Cancer Institute of Emory University, Atlanta, GA, USA; Emory Vaccine Center, Emory University, Atlanta, GA, USA; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
2
|
Chong AS, Habal M. Cutting to the chase: Pruning alloreactive T cells. Immunity 2025; 58:270-272. [PMID: 39938479 PMCID: PMC12121668 DOI: 10.1016/j.immuni.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 02/14/2025]
Abstract
Indirect CD4+ T cell allorecognition of donor peptides presented by host MHC class II antigens contributes to transplant rejection in part by eliciting donor-specific antibodies (DSAs). In this issue of Immunity, Zhanzak et al. revisit the role of indirectly alloreactive CD4+ T cells in transplantation and demonstrate that immunodominant epitopes stimulate a narrow repertoire of T cells that can be pruned to prevent DSA formation.
Collapse
Affiliation(s)
- Anita S Chong
- Department of Surgery, Section of Transplant, University of Chicago, Chicago, IL, USA.
| | - Marlena Habal
- Department of Medicine, Division of Cardiology, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
3
|
Yun IH, Yang J. Mechanisms of allorecognition and xenorecognition in transplantation. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:273-293. [PMID: 39743230 PMCID: PMC11732770 DOI: 10.4285/ctr.24.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025]
Abstract
Foreign antigen recognition is the ability of immune cells to distinguish self from nonself, which is crucial for immune responses in both invertebrates and vertebrates. In vertebrates, T cells play a pivotal role in graft rejection by recognizing alloantigens presented by antigen-presenting cells through direct, indirect, or semidirect pathways. B cells also significantly contribute to the indirect presentation of antigens to T cells. Innate immune cells, such as dendritic cells, identify pathogen- or danger-associated molecular patterns through pattern recognition receptors, thereby facilitating effective antigen presentation to T cells. Recent studies have shown that innate immune cells, including macrophages and NK cells, can recognize allogeneic or xenogeneic antigens using immune receptors like CD47 or activating NK receptors, instead of pattern recognition receptors. Additionally, macrophages and NK cells are capable of exhibiting memory responses to alloantigens, although these responses are shorter than those of adaptive memory. T cells also recognize xenoantigens through either direct or indirect presentation. Notably, macrophages and NK cells can directly recognize xenoantigens via surface immune receptors in an antibody-independent manner, or they can be activated in an antibody-dependent manner. Advances in our understanding of the recognition mechanisms of adaptive and innate immunity against allogeneic and xenogeneic antigens may improve our understanding of graft rejection.
Collapse
Affiliation(s)
- Il Hee Yun
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Jaeseok Yang
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Basu S, Dudreuilh C, Shah S, Sanchez-Fueyo A, Lombardi G, Dorling A. Activation and Regulation of Indirect Alloresponses in Transplanted Patients With Donor Specific Antibodies and Chronic Rejection. Transpl Int 2024; 37:13196. [PMID: 39228658 PMCID: PMC11368725 DOI: 10.3389/ti.2024.13196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
Following transplantation, human CD4+T cells can respond to alloantigen using three distinct pathways. Direct and semi-direct responses are considered potent, but brief, so contribute mostly to acute rejection. Indirect responses are persistent and prolonged, involve B cells as critical antigen presenting cells, and are an absolute requirement for development of donor specific antibody, so more often mediate chronic rejection. Novel in vitro techniques have furthered our understanding by mimicking in vivo germinal centre processes, including B cell antigen presentation to CD4+ T cells and effector cytokine responses following challenge with donor specific peptides. In this review we outline recent data detailing the contribution of CD4+ T follicular helper cells and antigen presenting B cells to donor specific antibody formation and antibody mediated rejection. Furthermore, multi-parametric flow cytometry analyses have revealed specific endogenous regulatory T and B subsets each capable of suppressing distinct aspects of the indirect response, including CD4+ T cell cytokine production, B cell maturation into plasmablasts and antibody production, and germinal centre maturation. These data underpin novel opportunities to control these aberrant processes either by targeting molecules critical to indirect alloresponses or potentiating suppression via exogenous regulatory cell therapy.
Collapse
Affiliation(s)
- Sumoyee Basu
- Centre for Nephrology, Urology and Transplantation, King’s College London, London, United Kingdom
- Department of Inflammation Biology, King’s College London, London, United Kingdom
| | - Caroline Dudreuilh
- Centre for Nephrology, Urology and Transplantation, King’s College London, London, United Kingdom
- Transplantation, Renal and Urology Directorate, Guy’s and St Thomas’ NHS Foundation Trust, Guy’s Hospital, London, United Kingdom
| | - Sapna Shah
- Centre for Nephrology, Urology and Transplantation, King’s College London, London, United Kingdom
- Renal Unit, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Alberto Sanchez-Fueyo
- Department of Inflammation Biology, King’s College London, London, United Kingdom
- Liver Sciences, King’s College London, London, United Kingdom
| | - Giovanna Lombardi
- Centre for Nephrology, Urology and Transplantation, King’s College London, London, United Kingdom
- Department of Inflammation Biology, King’s College London, London, United Kingdom
| | - Anthony Dorling
- Centre for Nephrology, Urology and Transplantation, King’s College London, London, United Kingdom
- Department of Inflammation Biology, King’s College London, London, United Kingdom
| |
Collapse
|
5
|
Carnel N, Lancia HH, Guinier C, Benichou G. Pathways of Antigen Recognition by T Cells in Allograft Rejection. Transplantation 2023; 107:827-837. [PMID: 36398330 PMCID: PMC10600686 DOI: 10.1097/tp.0000000000004420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The adaptive immune response leading to the rejection of allogeneic transplants is initiated and orchestrated by recipient T cells recognizing donor antigens. T-cell allorecognition is mediated via 3 distinct mechanisms: the direct pathway in which T cells recognize allogeneic major histocompatibility complex (MHC) molecules on donor cells, the indirect pathway through which T cells interact with donor peptides bound with self-MHC molecules on recipient antigen-presenting cells, and the recently described semidirect pathway whereby T cells recognize donor MHC proteins on recipient antigen-presenting cells. In this article, we present a description of each of these allorecognition pathways and discuss their role in acute and chronic rejection of allogeneic transplants.
Collapse
Affiliation(s)
- Natacha Carnel
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Hyshem H. Lancia
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Claire Guinier
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Gilles Benichou
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Hwang B, Bryers J, Mulligan MS. Potential role of exosome-based allorecognition pathways involved in lung transplant rejection. J Thorac Cardiovasc Surg 2020; 161:e129-e134. [PMID: 33258452 DOI: 10.1016/j.jtcvs.2020.04.183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/02/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023]
Abstract
Innate and adaptive immunity both contribute to allorecognition mechanisms that drive rejection after lung transplantation. Classic allorecognition pathways have been extensively described, but there continues to be several unanswered questions. Exosome research appears to be a novel and potentially significant area of allorecognition research and could be the missing link that answers some existing questions. This article reviews literature that is associated with allorecognition pathways and the role of exosomes in alloreactivity.
Collapse
Affiliation(s)
- Billanna Hwang
- Department of Surgery, University of Washington School of Medicine, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash; West Coast Exosortium (WestCo Exosortium), Seattle, Wash.
| | - James Bryers
- Center for Lung Biology, University of Washington, Seattle, Wash; West Coast Exosortium (WestCo Exosortium), Seattle, Wash; Department of Bioengineering, University of Washington, Seattle, Wash
| | - Michael S Mulligan
- Department of Surgery, University of Washington School of Medicine, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash; West Coast Exosortium (WestCo Exosortium), Seattle, Wash; Department of Medicine, University of Washington School of Medicine, Seattle, Wash
| |
Collapse
|
7
|
Li XG, Park IS, Choi BH, Kim UJ, Min BH. In Vivo Bioreactor Using Cellulose Membrane Benefit Engineering Cartilage by Improving the Chondrogenesis and Modulating the Immune Response. Tissue Eng Regen Med 2020; 17:165-181. [PMID: 32193874 PMCID: PMC7105552 DOI: 10.1007/s13770-019-00236-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND To regenerate tissue-engineered cartilage as a source of material for the restoration of cartilage defects, we used a human fetal cartilage progenitor cell pellet to improve chondrogenesis and modulation of the immune response in an in vivo bioreactor (IVB) system. METHODS IVB was buried subcutaneously in the host and then implanted into a cartilage defect. The IVB was composed of a silicone tube and a cellulose nano pore-sized membrane. First, fetal cartilage progenitor cell pellets were cultured in vitro for 3 days, then cultured in vitro, subcutaneously, and in an IVB for 3 weeks. First, the components and liquidity of IVB fluid were evaluated, then the chondrogenesis and immunogenicity of the pellets were evaluated using gross observation, cell viability assays, histology, biochemical analysis, RT-PCR, and Western blots. Finally, cartilage repair and synovial inflammation were evaluated histologically. RESULTS The fluid color and transparency of the IVB were similar to synovial fluid (SF) and the components were closer to SF than serum. The IVB system not only promoted the synthesis of cartilage matrix and maintained the cartilage phenotype, it also delayed calcification compared to the subcutaneously implanted pellets. CONCLUSION The IVB adopted to study cell differentiation was effective in preventing host immune rejection.
Collapse
Affiliation(s)
- Xue Guang Li
- Department of Orthopaedic Surgery, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea
- Cell Therapy Center, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - In-Su Park
- Cell Therapy Center, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - Byung Hyune Choi
- Department of Biomedical Sciences, Inha University College of Medicine, 100, Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Ung-Jin Kim
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 17104, Republic of Korea
- Department of Plant and Environmental New Resources, College of Life Sciences, Kyung Hee University, 1 Seocheon-dong, Giheung-gu, Yongin-si, Gyeonggi-do, 17104, Republic of Korea
| | - Byoung-Hyun Min
- Department of Orthopaedic Surgery, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea.
- Cell Therapy Center, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea.
- Department of Molecular Science and Technology, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea.
| |
Collapse
|
8
|
Geneugelijk K, Spierings E. PIRCHE-II: an algorithm to predict indirectly recognizable HLA epitopes in solid organ transplantation. Immunogenetics 2019; 72:119-129. [PMID: 31741009 PMCID: PMC6971131 DOI: 10.1007/s00251-019-01140-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/16/2019] [Indexed: 12/14/2022]
Abstract
Human leukocyte antigen (HLA) mismatches between donors and recipients may lead to alloreactivity after solid organ transplantation. Over the last few decades, our knowledge of the complexity of the HLA system has dramatically increased, as numerous new HLA alleles have been identified. As a result, the likelihood of alloreactive responses towards HLA mismatches after solid organ transplantation cannot easily be assessed. Algorithms are promising solutions to estimate the risk for alloreactivity after solid organ transplantation. In this review, we show that the recently developed PIRCHE-II (Predicted Indirectly ReCognizable HLA Epitopes) algorithm can be used to minimize alloreactivity towards HLA mismatches. Together with the use of other algorithms and simulation approaches, the PIRCHE-II algorithm aims for a better estimated alloreactive risk for individual patients and eventually an improved graft survival after solid organ transplantation.
Collapse
Affiliation(s)
- Kirsten Geneugelijk
- Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands.
| | - Eric Spierings
- Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| |
Collapse
|
9
|
Siu JHY, Surendrakumar V, Richards JA, Pettigrew GJ. T cell Allorecognition Pathways in Solid Organ Transplantation. Front Immunol 2018; 9:2548. [PMID: 30455697 PMCID: PMC6230624 DOI: 10.3389/fimmu.2018.02548] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 10/17/2018] [Indexed: 02/02/2023] Open
Abstract
Transplantation is unusual in that T cells can recognize alloantigen by at least two distinct pathways: as intact MHC alloantigen on the surface of donor cells via the direct pathway; and as self-restricted processed alloantigen via the indirect pathway. Direct pathway responses are viewed as strong but short-lived and hence responsible for acute rejection, whereas indirect pathway responses are typically thought to be much longer lasting and mediate the progression of chronic rejection. However, this is based on surprisingly scant experimental evidence, and the recent demonstration that MHC alloantigen can be re-presented intact on recipient dendritic cells-the semi-direct pathway-suggests that the conventional view may be an oversimplification. We review recent advances in our understanding of how the different T cell allorecognition pathways are triggered, consider how this generates effector alloantibody and cytotoxic CD8 T cell alloresponses and assess how these responses contribute to early and late allograft rejection. We further discuss how this knowledge may inform development of cellular and pharmacological therapies that aim to improve transplant outcomes, with focus on the use of induced regulatory T cells with indirect allospecificity and on the development of immunometabolic strategies. KEY POINTS Acute allograft rejection is likely mediated by indirect and direct pathway CD4 T cell alloresponses.Chronic allograft rejection is largely mediated by indirect pathway CD4 T cell responses. Direct pathway recognition of cross-dressed endothelial derived MHC class II alloantigen may also contribute to chronic rejection, but the extent of this contribution is unknown.Late indirect pathway CD4 T cell responses will be composed of heterogeneous populations of allopeptide specific T helper cell subsets that recognize different alloantigens and are at various stages of effector and memory differentiation.Knowledge of the precise indirect pathway CD4 T cell responses active at late time points in a particular individual will likely inform the development of alloantigen-specific cellular therapies and will guide immunometabolic modulation.
Collapse
|
10
|
Geneugelijk K, Spierings E. Matching donor and recipient based on predicted indirectly recognizable human leucocyte antigen epitopes. Int J Immunogenet 2018; 45:41-53. [PMID: 29464898 DOI: 10.1111/iji.12359] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/21/2017] [Accepted: 02/01/2018] [Indexed: 12/27/2022]
Abstract
The predicted indirectly recognizable human leucocyte antigen (HLA) epitopes (PIRCHE) algorithm is a novel in silico algorithm to determine donor-recipient compatibility. The PIRCHE algorithm determines donor-recipient compatibility by counting the number of mismatched HLA-derived epitopes that are involved in indirect T-cell alloimmune responses; these epitopes are designated as PIRCHE. Over the last few years, the PIRCHE algorithm has been investigated in both hematopoietic stem cell transplantation and solid organ transplantation. This review describes the theory of the algorithm, its application in transplantation, and highlights the future perspectives on the clinical application of the PIRCHE algorithm.
Collapse
Affiliation(s)
- K Geneugelijk
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - E Spierings
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
11
|
Abstract
The goal of cellular transplantation is to allow long-term function of the grafted cells using minimal host immunosuppression. To this end, the major strategies to implant cells and tissues are through: (i) the pretreatment of the graft to reduce tissue immunogenicity; (ii) the application of immunoisolation technologies to prevent host sensitization to implanted cells; and (iii) the induction of immunological tolerance to the donor tissues. Further, a major dilemma facing clinical tissue grafting is the shortage of donor tissue for transplantation. This problem requires the consideration of tissues from other species (xenografts) as a potential source of donor material. In light of these issues, the focus of this discussion is on the T cell-dependent response to allogeneic and xenogeneic transplants and the implications of this reactivity on the field of cellular replacement therapy.
Collapse
Affiliation(s)
- R G Gill
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Denver 80262, USA
| | | |
Collapse
|
12
|
Firl DJ, Benichou G, Kim JI, Yeh H. A Paradigm Shift on the Question of B Cells in Transplantation? Recent Insights on Regulating the Alloresponse. Front Immunol 2017; 8:80. [PMID: 28210263 PMCID: PMC5288351 DOI: 10.3389/fimmu.2017.00080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
B lymphocytes contribute to acute and chronic allograft rejection through their production of donor-specific antibodies (DSAs). In addition, B cells present allopeptides bound to self-MHC class II molecules and provide costimulation signals to T cells, which are essential to their activation and differentiation into memory T cells. On the other hand, both in laboratory rodents and patients, the concept of effector T cell regulation by B cells is gaining traction in the field of transplantation. Specifically, clinical trials using anti-CD20 monoclonal antibodies to deplete B cells and reverse DSA had a deleterious effect on rates of acute cellular rejection; a peculiar finding that calls into question a central paradigm in transplantation. Additional work in humans has characterized IL-10-producing B cells (IgM memory and transitional B cells), which suppress the proliferation and inflammatory cytokine productions of effector T cells in vitro. Understanding the mechanisms of regulating the alloresponse is critical if we are to achieve operational tolerance across transplantation. This review will focus on recent evidence in murine and human transplantation with respect to non-traditional roles for B cells in determining clinical outcomes.
Collapse
Affiliation(s)
- Daniel J Firl
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Gilles Benichou
- Transplant Center, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - James I Kim
- Transplant Center, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - Heidi Yeh
- Transplant Center, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
13
|
Abstract
The development of post-transplantation antibodies against non-HLA autoantigens is associated with rejection and decreased long-term graft survival. Although our knowledge of non-HLA antibodies is incomplete, compelling experimental and clinical findings demonstrate that antibodies directed against autoantigens such as angiotensin type 1 receptor, perlecan and collagen, contribute to the process of antibody-mediated acute and chronic rejection. The mechanisms that underlie the production of autoantibodies in the setting of organ transplantation is an important area of ongoing investigation. Ischaemia-reperfusion injury, surgical trauma and/or alloimmune responses can result in the release of organ-derived autoantigens (such as soluble antigens, extracellular vesicles or apoptotic bodies) that are presented to B cells in the context of the transplant recipient's antigen presenting cells and stimulate autoantibody production. Type 17 T helper cells orchestrate autoantibody production by supporting the proliferation and maturation of autoreactive B cells within ectopic tertiary lymphoid tissue. Conversely, autoantibody-mediated graft damage can trigger alloimmunity and the development of donor-specific HLA antibodies that can act in synergy to promote allograft rejection. Identification of the immunologic phenotypes of transplant recipients at risk of non-HLA antibody-mediated rejection, and the development of targeted therapies to treat such rejection, are sorely needed to improve both graft and patient survival.
Collapse
|
14
|
Vanikar AV, Trivedi HL, Kumar A, Gopal SC, Patel HV, Gumber MR, Kute VB, Shah PR, Dave SD. Co-infusion of donor adipose tissue-derived mesenchymal and hematopoietic stem cells helps safe minimization of immunosuppression in renal transplantation - single center experience. Ren Fail 2014; 36:1376-1384. [PMID: 25246338 DOI: 10.3109/0886022x.2014.950931] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Stem cell therapy (SCT) is used for immunosuppression minimization in renal transplantation (RT). We carried out a prospective study to evaluate the benefits of co-infusion of donor adipose-derived mesenchymal stem cells (AD-MSC) + hematopoietic stem cells (HSC) in living donor RT (LDRT) under non-myeloablative conditioning. METHODS In a demographically balanced three-armed LDRT trial with 95 patients in each arm, group-1 received portal co-infusion of AD-MSC + HSC, group-2 received HSC and group-3 received no SCT. Lymphoid irradiation and anti-thyroglobulin were used for conditioning. RESULTS SCT was safe. At 1 and 5 years post-transplant, patient survival was 100% and 94.7% in group-1, 100% and 95.7% in group-2, and 94.7% and 84% in group-3, death-censored graft survival was 100% and 94.6% in group-1, 100% and 91.3% in group-2, and 98.9% and 94.4% in group-3 with mean serum creatinine (mg/dL) of 1.38 and 1.39 in group-1, 1.48 and 1.51 in group-2, and 1.29 and 1.42 and in group-3. Rejection episodes and immunosuppression requirement were lesser in SCT groups versus controls with best results noted in group-1. CONCLUSION Coinfusion of donor AD-MSC +HSC in portal circulation pre-transplant under non-myeloablative conditioning is safe and effective for immunosuppression minimization in LDRT.
Collapse
Affiliation(s)
- Aruna V Vanikar
- Department of Pathology, Laboratory of Medicine, Transfusion Services and Immunohematology, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases & Research Centre (IKDRC), Dr. H.L. Trivedi Institute of Transplantation Sciences (ITS) , Ahmedabad, Gujarat , India
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Predicting alloreactivity in transplantation. J Immunol Res 2014; 2014:159479. [PMID: 24868561 PMCID: PMC4020392 DOI: 10.1155/2014/159479] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/13/2014] [Indexed: 01/10/2023] Open
Abstract
Human leukocyte Antigen (HLA) mismatching leads to severe complications after solid-organ transplantation and hematopoietic stem-cell transplantation. The alloreactive responses underlying the posttransplantation complications include both direct recognition of allogeneic HLA by HLA-specific alloantibodies and T cells and indirect T-cell recognition. However, the immunogenicity of HLA mismatches is highly variable; some HLA mismatches lead to severe clinical B-cell- and T-cell-mediated alloreactivity, whereas others are well tolerated. Definition of the permissibility of HLA mismatches prior to transplantation allows selection of donor-recipient combinations that will have a reduced chance to develop deleterious host-versus-graft responses after solid-organ transplantation and graft-versus-host responses after hematopoietic stem-cell transplantation. Therefore, several methods have been developed to predict permissible HLA-mismatch combinations. In this review we aim to give a comprehensive overview about the current knowledge regarding HLA-directed alloreactivity and several developed in vitro and in silico tools that aim to predict direct and indirect alloreactivity.
Collapse
|
16
|
Recognition of Foreign Antigen and Foreign Major Histocompatibility Complex. Xenotransplantation 2014. [DOI: 10.1128/9781555818043.ch9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
17
|
|
18
|
Kant CD, Akiyama Y, Tanaka K, Shea S, Connolly SE, Germana S, Winn HJ, LeGuern C, Tocco G, Benichou G. Primary vascularization of allografts governs their immunogenicity and susceptibility to tolerogenesis. THE JOURNAL OF IMMUNOLOGY 2013; 191:1948-56. [PMID: 23833234 DOI: 10.4049/jimmunol.1202092] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We investigated the influence of allograft primary vascularization on alloimmunity, rejection, and tolerance in mice. First, we showed that fully allogeneic primarily vascularized and conventional skin transplants were rejected at the same pace. Remarkably, however, short-term treatment of mice with anti-CD40L Abs achieved long-term survival of vascularized skin and cardiac transplants but not conventional skin grafts. Nonvascularized skin transplants triggered vigorous direct and indirect proinflammatory type 1 T cell responses (IL-2 and IFN-γ), whereas primarily vascularized skin allografts failed to trigger a significant indirect alloresponse. A similar lack of indirect alloreactivity was also observed after placement of different vascularized organ transplants, including hearts and kidneys, whereas hearts placed under the skin (nonvascularized) triggered potent indirect alloresponses. Altogether, these results suggest that primary vascularization of allografts is associated with a lack of indirect T cell alloreactivity. Finally, we show that long-term survival of vascularized skin allografts induced by anti-CD40L Abs was associated with a combined lack of indirect alloresponse and a shift of the direct alloresponse toward a type 2 cytokine (IL-4, IL-10)-secretion pattern but no activation/expansion of Foxp3(+) regulatory T cells. Therefore, primary vascularization of allografts governs their immunogenicity and tolerogenicity.
Collapse
Affiliation(s)
- Cavit D Kant
- Transplantation Unit and Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
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
|
20
|
Benichou G, Tonsho M, Tocco G, Nadazdin O, Madsen JC. Innate immunity and resistance to tolerogenesis in allotransplantation. Front Immunol 2012; 3:73. [PMID: 22566954 PMCID: PMC3342343 DOI: 10.3389/fimmu.2012.00073] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/22/2012] [Indexed: 01/06/2023] Open
Abstract
The development of immunosuppressive drugs to control adaptive immune responses has led to the success of transplantation as a therapy for end-stage organ failure. However, these agents are largely ineffective in suppressing components of the innate immune system. This distinction has gained in clinical significance as mounting evidence now indicates that innate immune responses play important roles in the acute and chronic rejection of whole organ allografts. For instance, whereas clinical interest in natural killer (NK) cells was once largely confined to the field of bone marrow transplantation, recent findings suggest that these cells can also participate in the acute rejection of cardiac allografts and prevent tolerance induction. Stimulation of Toll-like receptors (TLRs), another important component of innate immunity, by endogenous ligands released in response to ischemia/reperfusion is now known to cause an inflammatory milieu favorable to graft rejection and abrogation of tolerance. Emerging data suggest that activation of complement is linked to acute rejection and interferes with tolerance. In summary, the conventional wisdom that the innate immune system is of little importance in whole organ transplantation is no longer tenable. The addition of strategies that target TLRs, NK cells, complement, and other components of the innate immune system will be necessary to eventually achieve long-term tolerance to human allograft recipients.
Collapse
Affiliation(s)
- Gilles Benichou
- Transplant Research Center, Massachusetts General Hospital and Harvard Medical School Boston, MA, USA
| | | | | | | | | |
Collapse
|
21
|
Benichou G, Yamada Y, Yun SH, Lin C, Fray M, Tocco G. Immune recognition and rejection of allogeneic skin grafts. Immunotherapy 2012; 3:757-70. [PMID: 21668313 DOI: 10.2217/imt.11.2] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The transplantation of allogeneic skin grafts is associated with a potent inflammatory immune response leading to the destruction of donor cells and the rejection of the graft. Shortly after transplantation, skin dendritic cells (DCs) migrate out of the graft through lymphatic vessels and infiltrate the recipient's draining lymph nodes where they present donor antigens via two mechanisms: the direct pathway, in which T cells recognize intact donor MHC antigens on donor DCs; and the indirect pathway, involving T-cell recognition of donor peptides bound to self-MHC molecules on recipient DCs. Some recent studies have suggested that T cells can become activated via recognition of donor MHC molecules transferred on recipient antigen-presenting cells (semidirect pathway). Activation of T cells via direct or indirect allorecognition is sufficient to trigger acute rejection of allogeneic skin grafts. In addition, allospecific antibodies contribute to the rejection process either by killing allogeneic targets in a complement-dependent fashion or by opsonizing donor cells and forming immune complexes. Finally, several studies demonstrate that NK cells, activated due to missing self-MHC class I molecules on allogeneic cells, are involved in allogeneic skin graft rejection via direct killing of donor cells and through the production of proinflammatory cytokines including IFN-γ and TNF-α.
Collapse
Affiliation(s)
- Gilles Benichou
- Department of Surgery, Transplant Unit & Wellman Photomedicine Center Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Wendt H, Hillmer A, Reimers K, Kuhbier JW, Schäfer-Nolte F, Allmeling C, Kasper C, Vogt PM. Artificial skin--culturing of different skin cell lines for generating an artificial skin substitute on cross-weaved spider silk fibres. PLoS One 2011; 6:e21833. [PMID: 21814557 PMCID: PMC3144206 DOI: 10.1371/journal.pone.0021833] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 06/12/2011] [Indexed: 12/31/2022] Open
Abstract
Background In the field of Plastic Reconstructive Surgery the development of new innovative matrices for skin repair is in urgent need. The ideal biomaterial should promote attachment, proliferation and growth of cells. Additionally, it should degrade in an appropriate time period without releasing harmful substances, but not exert a pathological immune response. Spider dragline silk from Nephila spp meets these demands to a large extent. Methodology/Principal Findings Native spider dragline silk, harvested directly out of Nephila spp spiders, was woven on steel frames. Constructs were sterilized and seeded with fibroblasts. After two weeks of cultivating single fibroblasts, keratinocytes were added to generate a bilayered skin model, consisting of dermis and epidermis equivalents. For the next three weeks, constructs in co-culture were lifted on an originally designed setup for air/liquid interface cultivation. After the culturing period, constructs were embedded in paraffin with an especially developed program for spidersilk to avoid supercontraction. Paraffin cross- sections were stained in Haematoxylin & Eosin (H&E) for microscopic analyses. Conclusion/Significance Native spider dragline silk woven on steel frames provides a suitable matrix for 3 dimensional skin cell culturing. Both fibroblasts and keratinocytes cell lines adhere to the spider silk fibres and proliferate. Guided by the spider silk fibres, they sprout into the meshes and reach confluence in at most one week. A well-balanced, bilayered cocultivation in two continuously separated strata can be achieved by serum reduction, changing the medium conditions and the cultivation period at the air/liquid interphase. Therefore spider silk appears to be a promising biomaterial for the enhancement of skin regeneration.
Collapse
Affiliation(s)
- Hanna Wendt
- Department of Plastic, Hand, and Reconstructive Surgery, Medical School Hannover, Hannover, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Fernandes E, Goold HD, Kissenpfennig A, Malissen B, Dyson J, Bennett CL. The role of direct presentation by donor dendritic cells in rejection of minor histocompatibility antigen-mismatched skin and hematopoietic cell grafts. Transplantation 2011; 91:154-60. [PMID: 21085063 DOI: 10.1097/tp.0b013e318201ac27] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The success of transplantation is hampered by rejection of the graft by alloreactive T cells. Donor dendritic cells (DC) have been shown to be required for direct priming of immune responses to antigens from major histocompatibility complex-mismatched grafts. However, for immune responses to major histocompatibility complex-matched, minor histocompatibility (H) antigen mismatched grafts, the magnitude of the T-cell response to directly presented antigens is reduced, and the indirect pathway is more important. Therefore, we aimed to investigate the requirement for donor DC to directly present antigen from minor H antigen mismatched skin and hematopoietic grafts. METHODS Langerhans cell- or conventional (c)DC-depleted skin or hematopoietic cells from male DC-specific diphtheria toxin receptor mice were grafted onto, or injected into, syngeneic female recipients, and survival of the male tissue was compared with nondepleted tissue. Activation of the alloreactive immune response was tracked by the expansion of T cells specific for male HY-derived epitopes. RESULTS Our data demonstrate that depletion of donor Langerhans cell, dermal cDC, or both from skin grafts prolongs their survival but does not prevent rejection. Extended survival correlates with delayed expansion of HY peptide-specific CD8 T cells. In addition, depletion of donor cDC delays rejection of male hematopoietic cells. CONCLUSIONS Our results demonstrate for the first time that direct presentation of minor H antigens by donor DC is required for efficient rejection of skin and hematopoietic grafts by CD8 T cells. But, in the absence of donor DC, indirect presentation of minor antigens is sufficient to mediate the response.
Collapse
Affiliation(s)
- Eduardo Fernandes
- Section of Immunobiology, Division of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
24
|
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]
|
25
|
Issa F, Schiopu A, Wood KJ. Role of T cells in graft rejection and transplantation tolerance. Expert Rev Clin Immunol 2010; 6:155-69. [PMID: 20383898 DOI: 10.1586/eci.09.64] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Transplantation is the most effective treatment for end-stage organ failure, but organ survival is limited by immune rejection and the side effects of immunosuppressive regimens. T cells are central to the process of transplant rejection through allorecognition of foreign antigens leading to their activation, and the orchestration of an effector response that results in organ damage. Long-term transplant acceptance in the absence of immunosuppressive therapy remains the ultimate goal in the field of transplantation and many studies are exploring potential therapies. One promising cellular therapy is the use of regulatory T cells to induce a state of donor-specific tolerance to the transplant. This article first discusses the role of T cells in transplant rejection, with a focus on the mechanisms of allorecognition and the alloresponse. This is followed by a detailed review of the current progress in the field of regulatory T-cell therapy in transplantation and the translation of this therapy to the clinical setting.
Collapse
Affiliation(s)
- Fadi Issa
- Nuffield Department of Surgery, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | | | | |
Collapse
|
26
|
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
Affiliation(s)
- Behdad Afzali
- Department of Nephrology and Transplantation, Guy's Hospital, Kings College London, London, UK
| | | | | |
Collapse
|
27
|
Waanders MM, Heidt S, Koekkoek KM, Zoet YM, Doxiadis IIN, Amir A, Heemskerk MHM, Mulder A, Brand A, Roelen DL, Claas FHJ. Monitoring of indirect allorecognition: wishful thinking or solid data? ACTA ACUST UNITED AC 2008; 71:1-15. [PMID: 18096006 DOI: 10.1111/j.1399-0039.2007.00979.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Monitoring of T cells involved in the alloimmune response after transplantation requires the availability of reliable in vitro assays for the detection of T cells with both direct and indirect allospecificity. While generally accepted assays exist to measure helper and cytotoxic T cells involved in direct allorecognition, consensus about an assay for monitoring indirect T-cell allorecognition in clinical transplantation is lacking. Many studies claim a relationship between the reactivity of T cells with indirect allospecificity and graft rejection, but different protocols are used and essential controls are often lacking. In this review, the disadvantages and pitfalls of the current approaches are discussed, in some cases supported by the results of our own in vitro experiments. We conclude that an international workshop is necessary to establish and validate a uniform, robust and reliable assay for the monitoring of transplant recipients and to study the actual role of indirect allorecognition in acute and chronic rejection.
Collapse
Affiliation(s)
- M M Waanders
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Spadafora-Ferreira M, Caldas C, Faé KC, Marrero I, Monteiro SM, Lin-Wang HT, Socorro-Silva A, Fonseca SG, Fonseca JA, Kalil J, Coelho V. CD4+CD25+Foxp3+ indirect alloreactive T cells from renal transplant patients suppress both the direct and indirect pathways of allorecognition. Scand J Immunol 2007; 66:352-61. [PMID: 17635813 DOI: 10.1111/j.1365-3083.2007.01976.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alloreactive T cells recognize donor antigens by two routes: direct and indirect pathways of allorecognition. Although the direct pathway is reported to be dominant in allograft rejection, indirect allorecognition also plays an important role. Indirect alloreactivity is also observed in renal transplant patients irrespective of rejection. Previously we showed a predominance of interleukin (IL)-10 induced by indirect allorecognition of donor human leucocyte antigen (HLA)-DR peptides, suggesting the existence of indirect alloreactive T cells displaying regulatory activity. In the present work, our objective was to characterize these regulatory T cells. We detected indirect alloproliferation of peripheral blood mononuclear cells (PBMC) from renal transplant patients, induced by donor HLA-DR peptides, dependent on IL-4 or IL-10, suggesting regulatory activity as part of the alloreactive T-cell repertoire. PBMC-derived indirect alloreactive T-cell lines were established and produced both inflammatory and regulatory cytokines. We showed that two of these T-cell lines which were able to inhibit both direct and indirect alloproliferation of another T-cell line from the same patient presented a CD4(+)CD25(+)Foxp3(+) T-cell population. These data support the idea that indirect alloreactive T cells may also have regulatory activity and may contribute to the maintenance of the human renal allograft.
Collapse
Affiliation(s)
- M Spadafora-Ferreira
- Heart Institute (InCor), Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Archbold JK, Ely LK, Kjer-Nielsen L, Burrows SR, Rossjohn J, McCluskey J, Macdonald WA. T cell allorecognition and MHC restriction--A case of Jekyll and Hyde? Mol Immunol 2007; 45:583-98. [PMID: 17869342 DOI: 10.1016/j.molimm.2006.05.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Accepted: 05/19/2006] [Indexed: 01/14/2023]
Abstract
A great paradox in cellular immunology is how T cell allorecognition exists at high frequencies (up to 10%) despite the stringent requirements of discriminating 'self' from 'non-self' imposed by MHC restriction. Thus, in tissue transplantation, a substantial proportion of the recipient's T cells will have the ability to recognize the graft and instigate an immune response against the transplanted tissue, ultimately resulting in graft rejection--a manifestation of T cell alloreactivity. Transplantation of human organs and lymphoid cells as treatment for otherwise life-threatening diseases has become a more routine medical procedure making this problem of great importance. Immunologists have gained important insights into the mechanisms of T cell alloreactivity from cytotoxic T cell assays, affinity-avidity studies, and crystal structures of peptide-MHC (pMHC) molecules and T cell receptors (TCRs) both alone and in complex. Despite the clinical significance of alloreactivity, the crystal structure of an alloreactive human TCR in complex with both cognate pMHC and an allogeneic pMHC complex has yet to be determined. This review highlights some of the important findings from studies characterizing the way in which alloreactive T cell receptors and pMHC molecules interact in an attempt to resolve this great irony of the cellular immune response.
Collapse
Affiliation(s)
- Julia K Archbold
- The Protein Crystallography Unit, Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | | | |
Collapse
|
30
|
Semiletova NV, Shen XD, Feldman DM, Gao F, Mhoyan A, Liu D, Busuttil RW, Kupiec-Weglinski JW, Ghobrial RM. Class I MHC allochimeric presentation of composite immunogenic and self epitopes induces tolerance to genetically diverse rat strains. Cell Immunol 2007; 248:48-58. [PMID: 17936255 PMCID: PMC3699404 DOI: 10.1016/j.cellimm.2007.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2006] [Accepted: 04/16/2007] [Indexed: 10/22/2022]
Abstract
Functional topography of rat class I major histocompatibility complex (MHC) molecule was studied. The alpha1-helical sequences that are shared by class I RT1.A(l) and RT1.A(u) were substituted in the RT1.A(a) molecule to produce the composite [alpha(1h)(l/u)]-RT1.A(a) MHC class I allochimeric molecule. Dominant immunogenic epitopes that induce accelerated rejection were identified within the hypervariable regions of the alpha1 domain of RT1.A(a), RT1.A(l), and RT1.A(u). Peri-transplant portal venous delivery of MHC class I allochimeric proteins, that included composite alpha1 helical immunodominant epitopes of RT1.A(u) and RT1.A(l), induced donor-specific tolerance to RT1(u) (Wistar Furth, WF) and RT1(l) Lewis, LEW) disparate cardiac allografts in ACI (RT1(a)) hosts. Allochimeric generated tolerance was characterized by absence of T cell deletion or anergy. Donor specific IgM allo-Abs was not detected, while IgG alloresponse was markedly attenuated in sera of tolerant hosts. Further, long-term allografts in allochimeric-conditioned hosts exhibited moderate B cell infiltration when compared to rejecting controls. Analysis of intragraft cytokines revealed selective upregulation of IL-10 and marked inhibition of IL-2, IFN-gamma, and IL-4. Our findings indicate the emergence of a peripherally induced tolerant state, afforded by the novel approach of soluble class I allochimeric conditioning that presents donor immunogenic epitopes in the context of recipient class I determinants.
Collapse
Affiliation(s)
- Natalya V. Semiletova
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Xiu-Da Shen
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Daniel M. Feldman
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Feng Gao
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Ana Mhoyan
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Dhai Liu
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Ronald W. Busuttil
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jerzy W. Kupiec-Weglinski
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Rafik M. Ghobrial
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
31
|
Abstract
The inherent ability of the host immune system to distinguish between self- and non-self forms the basis of allorecognition. T lymphocytes constitute the most important effector arm of allorecognition. Here we describe the fundamentals of direct and indirect pathways by which allopeptides are presented to effector T cells. The nature of allopeptides presented along with tolerogenic strategies like altered peptide ligands and intra- or extra-thymic allopeptide inoculation are discussed. In addition, we speculate on the potential of regulatory T cells to modulate alloimmune responses.
Collapse
Affiliation(s)
- Ankit Bharat
- Department of Surgery, Washington University School of Medicine, Box 8109-3328 CSRB, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | | |
Collapse
|
32
|
Afzali B, Lechler RI, Hernandez-Fuentes MP. Allorecognition and the alloresponse: clinical implications. TISSUE ANTIGENS 2007; 69:545-56. [PMID: 17498264 DOI: 10.1111/j.1399-0039.2007.00834.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [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
|
33
|
Hanvesakul R, Maillere B, Briggs D, Baker R, Larché M, Ball S. Indirect recognition of T-cell epitopes derived from the alpha 3 and transmembrane domain of HLA-A2. Am J Transplant 2007; 7:1148-57. [PMID: 17355239 DOI: 10.1111/j.1600-6143.2007.01743.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Indirect allorecognition has been implicated in the mechanism of chronic rejection and alloantibody formation but precise definition of the epitopes involved has been limited. We have undertaken a detailed assessment of the antigenic properties of peptides derived from HLA-A2. Candidate epitopes were identified in vitro by assessment of MHC class II binding. The immune response to these epitopes was determined in patients awaiting a renal transplant by the assessment of PBMC activation using gamma-interferon ELISPOT. Twenty-two of fifty-five patients responded to peptides from HLA-A2 and this was associated with but not confined to those who had made antibody to HLA-A2 (14/18). Nineteen of twenty-two patients responded to peptides derived from the hypervariable alpha1 and alpha2 domains and 18/22 responded to peptides from the alpha 3 and transmembrane domain, the sequences of which show little polymorphism. In six patients, the sequence of these peptides was identical to self, that is, the response was autoimmune. The finding of indirect epitopes derived from regions of MHC class I that exhibit little polymorphism provides a novel perspective on the immune response to alloantigen and has potential implications for the development of specific therapies.
Collapse
Affiliation(s)
- R Hanvesakul
- Department of Renal Medicine, University Hospital Birmingham and Division of Medical Sciences, University of Birmingham, UK
| | | | | | | | | | | |
Collapse
|
34
|
Curry AJ, Pettigrew GJ, Negus MC, Easterfield AJ, Young JL, Bolton EM, Bradley JA. Dendritic cells internalise and re-present conformationally intact soluble MHC class I alloantigen for generation of alloantibody. Eur J Immunol 2007; 37:696-705. [PMID: 17266175 DOI: 10.1002/eji.200636543] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Following organ transplantation soluble MHC class I is released from the graft and may contribute to alloimmunity. We determined in a well-established rat model whether DC are able to internalise soluble MHC class I alloantigen and then re-present intact alloantigen to B cells and T cells for generation of an alloantibody or CD8 T cell response. PVG.RT1(u) BM-derived DC internalised (via an active process) and retained intact a recombinant soluble form of RT1-A(a) (sRT1-A(a)). When PVG.RT1(u) rats were immunised with sRT1-A(a)-pulsed syngeneic DC, they developed a strong anti-sRT1-A(a) alloantibody response and showed accelerated rejection of RT1-A(a)-disparate PVG.R8 heart grafts. Alloantibody production and accelerated heart graft rejection were both dependent on immunisation with viable sRT1-A(a)-pulsed DC. The alloantibody response to sRT1-A(a)-pulsed DC was directed exclusively against conformational epitopes expressed by sRT1-A(a) and not epitopes expressed, for example, by non-conformational sRT1-A(a) heavy chain. Immunisation with sRT1-A(a)-pulsed syngeneic DC did not stimulate a CD8 T cell response. Our findings suggest a novel alloantigen recognition pathway whereby soluble MHC class I alloantigen released from an allograft may be taken up by recipient DC and presented in an intact unprocessed form to B cells for the generation of an alloantibody response.
Collapse
Affiliation(s)
- Allison J Curry
- Department of Surgery, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Transplantation is an acceptable therapy for failing organs, however, the balance between prevention of acute rejection and immunosuppressant-induced toxicity remains elusive. Organ transplantation from a genetically disparate donor induces an immune response toward donor antigens in the recipient. An uncontrolled cumulative effect of these responses may jeopardize the recipient's life and destroy the grafted tissue. The donor antigen in the form of passenger leukocytes from the allograft migrating to the organized lymphoid collection is a prerequisite for initiation of acute rejection. In the host lymphoid tissue donor-specific dendritic cells primed with donor peptide activate naïve CD4 helper T cells which in turn activate effector CD8 T-cell clones through the release of cytokines. Activated effector CD8 cells return to the graft and augment destructive activity with the help of adhesive molecules and perforin. This seems to be the mechanism of adaptive immunity to destroy viral pathogens; the pattern of allograft injury is not much different. Adaptation and tolerance are based on the principle of exhaustion of donor-specific immune responses by an activation-deletion-exhaustion pathway.
Collapse
Affiliation(s)
- H L Trivedi
- Institute of Kidney Diseases and Transplantation Sciences, Gulabben Rasiklal Doshi and Kamlaben Mafatlal Mehta Institute, Research Center and Institute of Civil Hospital Campus, Asarwa Ahmedabad, Gujarat, India.
| |
Collapse
|
36
|
Abstract
Induction immunosuppression is intense, prophylactic therapy used at the time of transplantation based on the empiric observation that more powerful immunosuppression is required to prevent acute rejection early. In the past decade, there has been a growing trend towards the use of specialized agents such as antibody therapies for induction. In general, these agents have been shown to reduce the rate of acute rejection. However, their use has not been clearly shown to improve long-term transplant outcomes. This overview will review the biological basis for induction immunosuppression and the mechanisms of action of those specialized induction agents currently in clinical use. Clinical trials investigating induction regimens will be evaluated, and an individualized approach to the use of induction immunosuppressants will be presented.
Collapse
|
37
|
Kuo E, Maruyama T, Fernandez F, Mohanakumar T. Molecular mechanisms of chronic rejection following transplantation. Immunol Res 2006; 32:179-85. [PMID: 16106068 DOI: 10.1385/ir:32:1-3:179] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although significant advances have been made in the field of organ transplantation, chronic rejection remains a major limiting factor for prolonged graft survival. The long-term survival and function of transplanted lungs are limited by the development of bronchiolitis obliterans syndrome (BOS). The 10-yr lung graft survival rate is only 18.6%. Aside from results of several clinical studies that strongly support the concept that BOS results from alloimmune-mediated injury, little is known regarding specific immune effectors or target molecules involved in the pathogenesis of BOS. Studies from our laboratory have provided evidence for the seminal role of CD4+ T-cells in the pathogenesis of obliterative airway disease (OAD) seen in BOS. Prior to any clinically detectable lesions, there is indirect antigen presentation of mismatched major histocompatibility complex (MHC) class I antigen and production of antibodies to these MHC antigens. Both MHC and minor histocompatibility antigen disparities can result in the development of OAD in animal models and preliminary results strongly suggest that peptide vaccination strategies may prevent OAD following heterotopic tracheal transplants. Using a newly developed orthotopic tracheal transplant model, we have obtained evidence for an important and probably exclusive role for airway epithelial cell injury as a primary mechanism for the immunopathogenesis of the development of OAD.
Collapse
Affiliation(s)
- Elbert Kuo
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
38
|
Abstract
Dendritic cells (DCs) play a crucial role during the initiation of immune responses against non-self antigens. Following organ transplantation, activated donor- and recipient-derived DCs participate actively in graft rejection by sensitising recipient T cells via the direct or indirect pathways of allorecognition, respectively. There is increasing evidence that immature/semi-mature DCs induce antigen-specific unresponsiveness or tolerance to self antigens, both in central lymphoid tissue and in the periphery, through a variety of mechanisms (deletion, anergy and regulation). In the past few years, DC-based therapy of experimental allograft rejection has focused on ex vivo biological, pharmacological and genetic engineering of DCs to mimic/enhance their natural tolerogenicity. Successful outcomes in rodent models have built the case that DC-based therapy may provide a novel approach to transplant tolerance. Ongoing research into the role that DCs play in the induction of tolerance should allow for its clinical application in the near future.
Collapse
Affiliation(s)
- Mahyar Nouri-Shirazi
- Texas A&M University System Health Science Center, Baylor College of Dentistry, Department of Biomedical Sciences, Immunology Laboratory, 3302 Gaston Avenue, Dallas, TX 75246, USA.
| | | |
Collapse
|
39
|
Wallgren AC, Alder J, Andersson B, Karlsson-Parra A, Bäcker AE. The Direct Pathway of Human T-Cell Allorecognition is not Tolerized by Stimulation with Allogeneic Peripheral Blood Mononuclear Cells Irradiated with High-Dose Ultraviolet Ba. Scand J Immunol 2006; 63:90-6. [PMID: 16476007 DOI: 10.1111/j.1365-3083.2005.01720.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transfusions of high-dose (> or =10,000 Joule/m(2)) ultraviolet-B (UVB)-irradiated allogeneic leukocytes in rodent models have been shown to induce immunologic tolerance that is mediated by allospecific regulatory CD4(+) T cells. Whether these regulatory T cells recognize alloantigens through the direct or indirect pathway of allorecognition is controversial. Here, we demonstrate that the proliferative response obtained in standard primary mixed leukocyte reactions (MLRs) with human peripheral blood mononuclear cells (PBMCs) reflected a CD4(+) T-cell-dependent direct pathway of allorecognition and that high-dose UVB irradiation of PBMCs totally inhibited their capacity to induce a proliferative alloresponse. Re-stimulation with gamma-irradiated PBMCs from the same allogeneic donor (secondary MLR) elicited a proliferative and Th1-deviated response that was similar to the response induced in unprimed PBMCs. Finally, high-dose UVB was found to induce a rapid and massive apoptosis of irradiated PBMCs. Collectively, these data indicate that leukocytes irradiated with high-dose UVB are unable to prime for unresponsiveness or immune deviation in T cells directly recognizing allogeneic major histocompatibility complex molecules. Because it is well-established that antigens within transfused apoptotic cells are captured by resident tolerogenic spleen dendritic cells, we propose that tolerance induced by transfusions of high-dose UVB-irradiated leukocytes primarily involve T cells indirectly recognizing alloantigens.
Collapse
Affiliation(s)
- A C Wallgren
- Department of Endodontics and Oral Diagnostics, The Sahlgrenska Academy at Göteborg University, Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
40
|
Wallgren AC, Andersson B, Bäcker A, Karlsson-Parra A. Direct allorecognition promotes activation of bystander dendritic cells and licenses them for Th1 priming: a functional link between direct and indirect allosensitization. Scand J Immunol 2005; 62:234-42. [PMID: 16179010 DOI: 10.1111/j.1365-3083.2005.01663.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
T-cell sensitization to indirectly presented alloantigens (indirect pathway of allorecognition) plays a critical role in chronic rejection. The usual very efficient priming of such self-restricted, T helper type 1 (Th1)-deviated CD4+ T cells obviously conflicts with the fact that allogeneic MHC molecules are poorly immunogenic per se. The aim of the present study is to elucidate whether direct allosensitization induces production of inflammatory mediators that may affect recruitment and activation of immature bystander (host) dendritic cells (DC). These potential mechanisms were studied in vitro by conducting primary allogeneic mixed leucocyte reactions (MLR), mimicking the priming phase in secondary lymphoid organs, and secondary MLR, mimicking the effector phase within the graft. Primary, and particularly secondary, MLR supernatants were found to contain high levels of monocyte/immature DC-recruiting CC chemokines and pro-inflammatory cytokines. Exposure of immature DC to primary or secondary MLR supernatants was found to upregulate CD40 expression and further enhanced lipopolysaccharide-induced interleukin-12 (IL-12) p70 production. Secondary MLR supernatants additionally induced upregulation of CD86 and deviated allogeneic T-cell responses towards Th1 (enhanced interferon-gamma production without concomitant induction of detectable IL-4 or IL-10 production). These findings indicate that direct allorecognition may act as a Th1-deviating adjuvant for indirect allosensitization.
Collapse
Affiliation(s)
- A C Wallgren
- Department of Endodontics and Oral Diagnostics, The Sahlgrenska Academy at Göteborg University, Göteborg, Sweden.
| | | | | | | |
Collapse
|
41
|
Jiang S, Herrera O, Lechler RI. New spectrum of allorecognition pathways: implications for graft rejection and transplantation tolerance. Curr Opin Immunol 2005; 16:550-7. [PMID: 15341998 DOI: 10.1016/j.coi.2004.07.011] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It has long been appreciated that MHC alloantigens can be recognized via two pathways; direct and indirect. The relative contributions of these two pathways to transplant rejection are partially understood. In studies of transplantation tolerance it appears that regulatory T cells (Trs) with indirect allospecificity, particularly the CD4+CD25+ population, play a key role and can regulate responder cells with direct allospecificity for the same alloantigens. One of the conundrums that remains is how helper T and Tr cells with indirect allospecificity regulate T cells with direct allospecificity. At face value, this appears to break the rules of linkage that require interacting T cells to make contact with the same antigen-presenting cell. A third, 'semi-direct' pathway involving MHC exchange may help to resolve this conundrum. Insights into how these pathways interact in transplant immunity and tolerance will assist the pursuit of clinical tolerance.
Collapse
Affiliation(s)
- Shuiping Jiang
- Department of Immunology, Faculty of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, W120NN, UK.
| | | | | |
Collapse
|
42
|
Cortesini NSF, Colovai AI, Manavalan JS, Galluzzo S, Naiyer AJ, Liu J, Vlad G, Kim-Schulze S, Scotto L, Fan J, Cortesini R. Role of regulatory and suppressor T-cells in the induction of ILT3+ ILT4+ tolerogenic endothelial cells in organ allografts. Transpl Immunol 2005; 13:73-82. [PMID: 15380537 DOI: 10.1016/j.trim.2004.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Accepted: 06/03/2004] [Indexed: 11/16/2022]
|
43
|
Tokita D, Ohdan H, Onoe T, Hara H, Tanaka Y, Asahara T. Liver sinusoidal endothelial cells are insufficient to activate T cells. Transpl Int 2004; 18:237-45. [PMID: 15691278 DOI: 10.1111/j.1432-2277.2004.00045.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver sinusoidal endothelial cells (LSEC) have been reported to express MHC class II, CD80, CD86, and CD11c and effectively stimulate naive T cells. Because dendritic cells (DC) are known to possess these characteristics, we sought to directly compare the phenotype and function of murine LSEC and DC. Nonparenchymal cells from C57BL/6 mice were obtained by collagenase digestion of the liver followed by density gradient centrifugation. From the enriched nonparenchymal cell fraction, LSEC (CD45(-)) were then isolated to 99% purity using immunomagnetic beads. Flow cytometric analysis of LSEC demonstrated high expression of CD31, von Willebrand factor, and FcgammaRs. However, unlike DC, LSEC had low or absent expression of MHC class II, CD86, and CD11c. LSEC demonstrated a high capacity for Ag uptake in vitro and in vivo. Although acetylated low-density lipoprotein uptake has been purported to be a specific function of LSEC, we found DC captured acetylated low-density lipoprotein to a similar extent in vivo. Consistent with their phenotype, LSEC were poor stimulators of allogeneic T cells. Furthermore, in the absence of exogenous costimulation, LSEC induced negligible proliferation of CD4(+) or CD8(+) TCR-transgenic T cells. Thus, contrary to previous reports, our data indicate that LSEC alone are insufficient to activate naive T cells.
Collapse
Affiliation(s)
- Daisuke Tokita
- Department of Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi, Minami-Ku, Hiroshima, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Sauvé D, Baratin M, Leduc C, Bonin K, Daniel C. Alloantibody production is regulated by CD4+ T cells' alloreactive pathway, rather than precursor frequency or Th1/Th2 differentiation. Am J Transplant 2004; 4:1237-45. [PMID: 15268724 DOI: 10.1111/j.1600-6143.2004.00520.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Although CD4(+) T cells play an important role in the regulation of allograft rejection, the exact mechanisms by which they operate and the actual contribution of direct and indirect alloreactivity pathways remain to be fully characterized. Previous studies have established a possible relationship between the indirect alloreactivity pathway and antibody production, but interpretation of these results have been complicated by shortcomings inherent to the models used in these studies. To address this issue, we have developed a model based on TCR transgenic mice derived from a CD4(+) T-cell clone which recognize specific alloantigens by both alloreactivity pathways. Skin allografts on alphabeta T-cell deficient mice adoptively transferred with transgenic CD4(+) T cells were rejected without significant delay between the two alloreactivity pathways. No IgG alloantibody was produced following allograft rejection by the direct alloreactivity pathway alone. Importantly, production of antibodies against alloantigens of the direct pathway was shown to require help from CD4(+) T cells activated by the indirect pathway. These results indicate that the events leading to the initiation of immune responses responsible for graft rejection are clearly dependent on the population of antigen-presenting cells involved in T- and B-lymphocyte activation.
Collapse
Affiliation(s)
- Dominike Sauvé
- Institut National de la Recherche Scientifique-Institut Armand-Frappier, Université du Québec, Laval, Québec, Canada
| | | | | | | | | |
Collapse
|
45
|
Reed AJ, Noorchashm H, Rostami SY, Zarrabi Y, Perate AR, Jeganathan AN, Caton AJ, Naji A. Alloreactive CD4 T cell activation in vivo: an autonomous function of the indirect pathway of alloantigen presentation. THE JOURNAL OF IMMUNOLOGY 2004; 171:6502-9. [PMID: 14662850 DOI: 10.4049/jimmunol.171.12.6502] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation of alloreactive CD4 T cells occurs via the direct and indirect pathways of alloantigen presentation. A novel TCR/alloantigen transgenic system was designed that permitted in vivo visualization of CD4 T cell priming through these pathways. When both pathways of alloantigen presentation were intact, CD4 T cell activation in response to cardiac allografts was rapid and systemic by day 4 after transplantation, in contrast to that seen in response to skin allografts, which was delayed until 10-12 days after transplantation. Despite this systemic CD4 T cell activation in response to cardiac allografts, there was a paucity of activated graft-infiltrating CD4 T cells at 4 days posttransplantation. This finding suggests that the initial priming of alloimmune CD4 T cell responses occurs within draining lymphoid organs. Furthermore, alloantigens derived from cardiac allografts failed to promote thymic negative selection of developing thymocytes expressing the alloreactive TCR clonotype. In the absence of a functional direct pathway, the kinetics of activation, anatomic localization, and effector function of alloreactive CD4 T cells remained unchanged. Overall, the present study defines the anatomic and temporal characteristics of CD4 T cell alloimmune responses and demonstrates that CD4 T cell priming via the indirect pathway proceeds optimally in the absence of the direct pathway of alloantigen presentation.
Collapse
Affiliation(s)
- Amy J Reed
- Harrison Department of Surgical Research, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Xu B, Sakkas LI, Goldman BI, Jeevanandam V, Gaughan J, Oleszak EL, Platsoucas CD. Identical alpha-chain T-cell receptor transcripts are present on T cells infiltrating coronary arteries of human cardiac allografts with chronic rejection. Cell Immunol 2004; 225:75-90. [PMID: 14698142 DOI: 10.1016/j.cellimm.2003.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Chronic cardiac allograft rejection is characterized by graft arteriopathy and is a major obstacle of graft survival. We investigated T-cell receptor (TCR) alpha-chain transcripts of T cells infiltrating human epicardial coronary arteries from cardiac allografts with chronic rejection. The non-palindromic adaptor-polymerase chain reaction (NPA-PCR) was used to specifically amplify TCR alpha-chain transcripts from five explanted cardiac allografts with chronic rejection. The amplified products were cloned and sequenced to obtain the entire ValphaJalpha region. Immuno-histochemistry was used to identify the mononuclear cell infiltrates in the coronary arteries. All the five coronary artery specimens exhibited large populations of infiltrating mononuclear cells, which were primarily comprised of T cells and macrophages. In three specimens, high proportions ( approximately 80%) of identical alpha-chain TCR transcripts were detected. In peripheral blood mononuclear cells from a healthy individual, alpha-chain TCR transcripts were unique when compared to each other. Endomyocardial biopsies collected from one patient six months before the allograft was explanted, contained identical alpha-chain TCR transcripts to those found to be clonally expanded in the coronary arteries from this patient. These results indicate that T cells infiltrating the epicardial arteries of cardiac allografts with chronic rejection undergo proliferation and clonal expansion in response to a specific antigen, which very likely is an (allo)antigen(s).
Collapse
Affiliation(s)
- Bin Xu
- Department of Microbiology and Immunology, The Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Habiro K, Kotani M, Omoto K, Kobayashi S, Tanabe K, Shimmura H, Suzuki K, Hayashi T, Toma H, Abe R. Mechanism of allorecognition and skin graft rejection in CD28 and CD40 ligand double-deficient mice. Transplantation 2003; 76:854-8. [PMID: 14501867 DOI: 10.1097/01.tp.0000084868.09385.83] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND It has been shown that simultaneous blockade of CD28- and CD40-mediated costimulatory signals significantly prolongs allograft survival. Although these results led to an expectation of the establishment of specific immunotolerant therapy for organ transplantation, it became evident that these treatments rarely resulted in indefinite allograft survival. To uncover the mechanisms underlying these costimulation blockade-resistant allograft rejections, we studied the process of allogenic skin graft rejection in CD28 and CD40 ligand (L) double-deficient (double-knockout [dKO]) mice. METHODS Skin grafts from BALB/c or BALB.B mice were transplanted to C57BL/6 background dKO mice. The frequency of CD4+ and CD8+ T cells responding to alloantigens presented by direct or indirect pathways were defined by the use of a cytostaining assay. RESULTS BALB/c skin grafts were rapidly rejected by dKO mice. This CD28 and CD40L independent allograft rejection was inhibited by the depletion of CD8+ T cells. In vitro studies indicated that CD8+ T cells from BALB/c skin-grafted dKO mice responded to donor antigen presented only by the direct pathway. Unlike major histocompatibility complex (MHC)-mismatched donors, allogenic skin grafts from MHC-matched donors were accepted by dKO mice. CONCLUSION In the absence of CD28 and CD40 costimulatory signals, CD8+ T cells recognize MHC antigens by the direct pathway, resulting in the rejection of skin grafts from MHC-mismatched donors. In contrast, MHC-matched and non-MHC-mismatched donor skin grafts indefinitely survive in dKO mice. These results indicated that donor-host MHC matching may still be critical to costimulation blockade therapy for organ transplantation.
Collapse
Affiliation(s)
- Katsuyoshi Habiro
- Division of Immunobiology, Research Institute for Biological Sciences, Tokyo University of Science, Noda City, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mhoyan A, Wu GD, Kakoulidis TP, Que X, Yolcu ES, Cramer DV, Shirwan H. Predominant expression of the Th2 response in chronic cardiac allograft rejection. Transpl Int 2003. [DOI: 10.1111/j.1432-2277.2003.tb00350.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
49
|
Shirwan H, Mhoyan A, Kakoulidis TP, Yolcu ES, Ibrahim S. Prevention of chronic rejection with immunoregulatory cells induced by intrathymic immune modulation with class I allopeptides. Am J Transplant 2003; 3:581-9. [PMID: 12752314 DOI: 10.1034/j.1600-6143.2003.00052.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Intrathymic immune modulation with RT1.Aa allopeptides in the PVG.R8-to-PVG.1 U rat strain combination leads to long-term survival of cardiac allografts. This regimen, however, does not induce transplantation tolerance, since most long-surviving allografts undergo chronic rejection. We investigated recipients with chronic rejection for donor-specific immune nonresponsiveness and immunoregulatory cells as possible mechanisms responsible for long-term graft survival. There was a significant reduction in the proliferative response of T cells from long-term allograft recipients to donor alloantigens as compared with that of naïve T cells. Adoptive transfer of splenocytes from intrathymically manipulated primary long-term graft survivors into minimally irradiated secondary hosts resulted in indefinite survival of > 80% of allografts, providing evidence for immunoregulatory cells. Secondary recipients had total absence of donor-reactive cellular and humoral responses. Immunoregulation was also transferable from secondary to tertiary graft recipients. More importantly, there was a significant reduction in the incidence of chronic rejection in secondary hosts (> 85%) and complete prevention of acute and chronic rejection in tertiary hosts. This study demonstrates that intrathymic immunomodulation with class I allopeptides results in the generation of immunoregulatory cells that do not block chronic rejection in primary hosts where they develop, but prevent both acute and chronic allograft rejection when adoptively transferred into secondary and tertiary recipients.
Collapse
Affiliation(s)
- Haval Shirwan
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky 40202, USA.
| | | | | | | | | |
Collapse
|
50
|
Chalermskulrat W, Neuringer IP, Brickey WJ, Felix NJ, Randell SH, Ting JP, Aris RM. Hierarchical contributions of allorecognition pathways in chronic lung rejection. Am J Respir Crit Care Med 2003; 167:999-1007. [PMID: 12446274 DOI: 10.1164/rccm.200209-1099oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The role of allorecognition in initiating lung graft rejection is not clearly defined. Using the heterotopic tracheal transplantation model, we examined the contributions of the indirect and direct allorecognition pathways in chronic airway rejection. Fully mismatched, wild-type grafts were transplanted into major histocompatibility complex (MHC) II-/-, class II-like accessory molecule (H2-DMalpha)-/- using MHC I-/- and wild-type allorecipients as control subjects. Similarly, MHC I-/-, MHC II-/-, or MHC I/II-/- allografts were transplanted into wild-type mice with appropriate control subjects. Grafts from nonimmunosuppressed recipients were evaluated at Weeks 2, 4, and 6. Grafts transplanted into MHC II-/- and H2-DMalpha-/- allorecipients showed a more intact epithelium and reduced lumen obliteration compared with grafts transplanted into wild-type or MHC I-/- allorecipients (p < 0.05 for each). These grafts exhibited abundant CD4+ and CD8+ cell infiltrates similar to control allografts. MHC I-/- and MHC I/II-/- but not MHC II-/- allografts placed in wild-type animals demonstrated less severe rejection compared with allograft control subjects (p < 0.05 for each). Although the indirect allorecognition pathway has the strongest influence on rejection, the direct pathway is sufficient to ultimately cause chronic airway rejection. In addition, these results suggest that MHC class I molecules are the principal alloantigens in the mouse heterotopic tracheal model of obliterative bronchiolitis.
Collapse
Affiliation(s)
- Worakij Chalermskulrat
- Division of Pulmonary Disease and Critical Care Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|