1
|
Pettigrew GJ. Direct and indirect allorecognition-not so different after all? Am J Transplant 2025; 25:893-895. [PMID: 39993570 DOI: 10.1016/j.ajt.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Affiliation(s)
- Gavin J Pettigrew
- Department of Surgery, University of Cambridge, Hills Road, Cambridge CB2 0QQ, United Kingdom.
| |
Collapse
|
2
|
Benichou G, Lancia HH. Intercellular transfer of MHC molecules in T cell alloimmunity and allotransplantation. Biomed J 2024; 47:100749. [PMID: 38797478 PMCID: PMC11414654 DOI: 10.1016/j.bj.2024.100749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024] Open
Abstract
After transplantation of allogeneic tissues and organs, recognition by recipient T cells of donor MHC molecules initiates the pro-inflammatory adaptive immune response leading to allograft rejection. T cell allorecognition has long been known to be mediated via two distinct pathways: the direct pathway in which T cells recognize intact allogeneic MHC molecules displayed on donor cells and the indirect pathway whereby T cells recognize donor MHC peptides processed and presented by recipient antigen-presenting cells (APCs). It is believed that direct allorecognition is the driving force behind early acute allograft rejection while indirect allorecognition is involved in chronic allograft rejection, a progressive condition characterized by graft vasculopathy and tissue fibrosis. Recently, we and others have reported that after transplantation of allogeneic skin and organs, donor MHC molecules are transferred from donor cells to the host's APCs via trogocytosis or extracellular vesicles. Recipient APCs having captured donor MHC molecules can either present them to T cells in their intact form on their surface (semi-direct pathway) or the form of peptides bound to self-MHC molecules (indirect pathway). The present article provides an overview of recent studies evaluating the role of intercellular exchange of MHC molecules in T cell alloimmunity and its contribution to allograft rejection and tolerance.
Collapse
Affiliation(s)
- Gilles Benichou
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, MA, USA.
| | - Hyshem H Lancia
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, MA, USA
| |
Collapse
|
3
|
DiToro D, Murakami N, Pillai S. T-B Collaboration in Autoimmunity, Infection, and Transplantation. Transplantation 2024; 108:386-398. [PMID: 37314442 PMCID: PMC11345790 DOI: 10.1097/tp.0000000000004671] [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: 06/15/2023]
Abstract
We have attempted here to provide an up-to-date review of the collaboration between helper T cells and B cells in response to protein and glycoprotein antigens. This collaboration is essential as it not only protects from many pathogens but also contributes to a litany of autoimmune and immune-mediated diseases.
Collapse
Affiliation(s)
- Daniel DiToro
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Naoka Murakami
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Shiv Pillai
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
| |
Collapse
|
4
|
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
|
5
|
Farshbafnadi M, Razi S, Rezaei N. Transplantation. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
6
|
Hu JL, Kim BJ, Yu NH, Kwon ST. Impact of Injection Frequency of Adipose-Derived Stem Cells on Allogeneic Skin Graft Survival Outcomes in Mice. Cell Transplant 2021; 30:9636897211041966. [PMID: 34538121 PMCID: PMC8743972 DOI: 10.1177/09636897211041966] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Previous studies indicated that mesenchymal stem cells (MSCs) exhibit immunomodulatory properties in composite tissue allotransplantation. However, due to the high immunogenicity of skin, although the single administration of MSCs improves survival of the skin allotransplant, immune rejection is still inevitable. The aim of our study was to evaluate whether multiple administrations of MSCs would improve immune tolerance in the allogeneic skin graft, compared to that with a single administration in a mouse model. After full-thickness skin allotransplantation on the backs of the mice, the recipient mice were infused with phosphate-buffered saline and isogenic 1.5 × 105/mL adipose-derived stem cells (ADSCs). ADSCs were transplanted into different mice according to the different injection frequencies such as single, once a week, and twice a week. Skin sections were taken on days 7 and 21 post-transplantation in all groups for gene expression and histological studies. ADSCs increased skin allograft survival compared to that in control mice (P < 0.05). Interleukin-6 and tumor necrosis factor-alpha messenger RNA levels were decreased, and the abundance of lymphocytes, based on immunohistochemistry, was also decreased in ADSC-infused mice (P < 0.05). However, among the different ADSC injection frequency groups, multiple ADSC infusion did not improve the survival rate and decreased proinflammatory cytokines and lymphocytes, compared to those with the single administration of ADSCs (P > 0.05). Conversely, the results with single administration were slightly better than those with multiple administrations. Our study demonstrated that ADSCs have the potential for immunomodulation in vivo. However, the results with multiple ADSC administration were not as good as those with single administration, which indicates the complexity of ADSCs in vivo and implying the need for adequate preclinical experimentation.
Collapse
Affiliation(s)
- Ju Long Hu
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Byung Jun Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Na Hee Yu
- Biomedical Research Institute, Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Tack Kwon
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Xu Z, Lin CC, Ho S, Vlad G, Suciu-Foca N. Suppression of Experimental Autoimmune Encephalomyelitis by ILT3.Fc. THE JOURNAL OF IMMUNOLOGY 2020; 206:554-565. [PMID: 33361206 DOI: 10.4049/jimmunol.2000265] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 11/25/2020] [Indexed: 01/29/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the CNS that is characterized by demyelination, axonal loss, gliosis, and inflammation. The murine model of MS is the experimental autoimmune encephalopathy (EAE) induced by immunization of mice with myelin oligodendrocyte glycoprotein (MOG)35-55 Ig-like transcript 3 (ILT3) is an inhibitory cell surface receptor expressed by tolerogenic human dendritic cells. In this study, we show that the recombinant human ILT3.Fc protein binds to murine immune cells and inhibits the release of proinflammatory cytokines that cause the neuroinflammatory process that result in paralysis. Administration of ILT3.Fc prevents the rapid evolution of the disease in C57BL/6 mice and is associated with a profound reduction of proliferation of MOG35-55-specific Th1 and Th17 cells. Inhibition of IFN-γ and IL-17A in mice treated with ILT3.Fc is associated with delayed time of onset of the disease and its evolution to a peak clinical score. Neuropathological analysis shows a reduction in inflammatory infiltrates and demyelinated areas in the brains and spinal cords of treated mice. These results indicate that inhibition of Th1 and Th17 development provides effective suppression of EAE and suggests the feasibility of a clinical approach based on the use of ILT3.Fc for treatment of MS. Furthermore, our results open the way to further studies on the effect of the human ILT3.Fc protein in murine experimental models of autoimmunity and cancer.
Collapse
Affiliation(s)
- Zheng Xu
- Division of Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; and
| | - Chun-Chieh Lin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Sophey Ho
- Division of Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; and
| | - George Vlad
- Division of Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; and
| | - Nicole Suciu-Foca
- Division of Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; and
| |
Collapse
|
8
|
Karahan GE, Claas FHJ, Heidt S. Pre-existing Alloreactive T and B Cells and Their Possible Relevance for Pre-transplant Risk Estimation in Kidney Transplant Recipients. Front Med (Lausanne) 2020; 7:340. [PMID: 32793610 PMCID: PMC7385137 DOI: 10.3389/fmed.2020.00340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/08/2020] [Indexed: 12/25/2022] Open
Abstract
In allogeneic transplantation, genetic disparities between patient and donor may lead to cellular and humoral immune responses mediated by both naïve and memory alloreactive cells of the adaptive immune system. This review will focus on alloreactive T and B cells with emphasis on the memory compartment, their role in relation to kidney rejection, and in vitro assays to detect these alloreactive cells. Finally, the potential additional value of utilizing donor-specific memory T and B cell assays supplementary to current routine pre-transplant risk assessment of kidney transplant recipients will be discussed.
Collapse
Affiliation(s)
- Gonca E Karahan
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Sebastiaan Heidt
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
9
|
Subbotin VM. Pattern of organ remodeling in chronic non-communicable diseases is due to endogenous regulations and falls under the category of Kauffman's self-organization: A case of arterial neointimal pathology. Med Hypotheses 2020; 143:110106. [PMID: 32759005 DOI: 10.1016/j.mehy.2020.110106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/07/2020] [Accepted: 07/11/2020] [Indexed: 01/10/2023]
Abstract
Clinical diagnosis is based on analysis of pathologic findings that may result in perceived patterns. The same is true for diagnostic pathology: Pattern analysis is a foundation of the histopathology-based diagnostic system and, in conjunction with clinical and laboratory findings, forms a basis for the classification of diseases. Any histopathology diagnosis is based on the explicit assumption that the same diseased condition should result in formation of the same (or highly similar) morphologic patterns in different individuals; it is a standard approach in microscopic pathology, including that of non-communicable chronic diseases with organ remodeling. During fifty years of examining diseased tissues under microscopy, I keep asking the same question: Why is a similarity of patterns expected for chronic organ remodeling? For infection diseases, xenobiotic toxicity and deficiencies forming an identical pathologic pattern in different individuals is understandable and logical: The same infection, xenobiotic, or deficiency strikes the same target, which results in identical pathology. The same is true for Mendelian diseases: The same mutations lead to the same altered gene expressions and the same pathologic pattern. But why does this regularity hold true for chronic diseases with organ remodeling? Presumable causes (or risk factors) for a particular chronic disease differ in magnitude and duration between individuals, which should result in various series of transformations. Yet, mysteriously enough, pathological remodeling in a particular chronic disease always falls into a main dominating pattern, perpetuating and progressing in a similar fashion in different patients. Furthermore, some chronic diseases of different etiologies and dissimilar causes/risk factors manifest as identical or highly similar patterns of pathologic remodeling. HYPOTHESIS: I hypothesize that regulations governing a particular organ's chronic remodeling were selected in evolution as the safest response to various insults and physiologic stress conditions. This hypothesis implies that regulations directing diseased chronic remodeling always preexist but normally are controlled; this control can be disrupted by a diverse range of non-specific signals, liberating the pathway for identical pathologic remodeling. This hypothesis was tested in an analysis of arterial neointimal formation, the identical pathology occurring in different diseases and pathological conditions: graft vascular disease in organ transplantation, in-stent restenosis, peripheral arterial diseases, idiopathic intimal hyperplasia, Kawasaki disease, coronary atherosclerosis and as reaction to drugs. The hypothesis suggests that arterial intimal cells are poised between only two alternative pathways: the pathway with controlled intimal cell proliferation or the pathway where such control is disrupted, ultimately leading to the progressive neointimal pathology. By this property the arterial neointimal formation constitutes a special case of Kauffman's self-organization. This new hypothesis gives a parsimonious explanation for identical pathological patterns of arterial remodeling (neointimal formation), which occurs in diseases of different etiologies and due to dissimilar causes/risk factors, or without any etiology and causes/risk factors at all. This new hypothesis also suggests that regulation facilitating intimal cell proliferation cannot be overwritten or annulled because this feature is vital for arterial differentiation, cell renewal, and integrity. This hypothesis suggests that studying numerous, and likely interchangeable, non-specific signals that disrupt regulation controlling intimal cell proliferation is unproductive; instead, a study of the controlling regulation(s) itself should be a priority of our research.
Collapse
Affiliation(s)
- Vladimir M Subbotin
- University of Pittsburgh, Pittsburgh, PA 15260, USA; University of Wisconsin, Madison, WI 53705, USA; Arrowhead Parmaceuticals, Madison, WI 53719, USA.
| |
Collapse
|
10
|
Brossart P. The Role of Antigen Spreading in the Efficacy of Immunotherapies. Clin Cancer Res 2020; 26:4442-4447. [PMID: 32357962 DOI: 10.1158/1078-0432.ccr-20-0305] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/17/2020] [Accepted: 04/29/2020] [Indexed: 11/16/2022]
Abstract
The introduction and the unexpected efficacy of checkpoint inhibitors (CPI) and more recently of chimeric antigen receptor T cells (CAR T-cells) in the treatment of malignant diseases boosted the efforts in the development and clinical application of immunotherapeutic approaches. However, the definition of predictive factors associated with clinical responses as well as the identification of underlying mechanisms that promote the therapeutic efficacy remain to be determined. Starting from the first immunotherapeutic trials, it became evident that vaccine-induced tumor-specific T cells or the adoptive transfer of ex vivo-expanded T lymphocytes can recognize and eliminate malignant cells leading to long-lasting remissions in some patients. In addition, a phenomenon called epitope spreading, which was observed in responding patients, seemed to increase the efficiency possibly representing an important predictive factor. This review will focus on experimental and clinical evidence for the induction of epitope spreading and its role in the maintenance of an efficient antitumor immune response in cancer immunotherapy.
Collapse
Affiliation(s)
- Peter Brossart
- Department of Oncology, Haematology, Immuno-Oncology and Rheumatogy, University of Bonn, Bonn, Germany.
| |
Collapse
|
11
|
Lo MW, Woodruff TM. Complement: Bridging the innate and adaptive immune systems in sterile inflammation. J Leukoc Biol 2020; 108:339-351. [PMID: 32182389 DOI: 10.1002/jlb.3mir0220-270r] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The complement system is a collection of soluble and membrane-bound proteins that together act as a powerful amplifier of the innate and adaptive immune systems. Although its role in infection is well established, complement is becoming increasingly recognized as a key contributor to sterile inflammation, a chronic inflammatory process often associated with noncommunicable diseases. In this context, damaged tissues release danger signals and trigger complement, which acts on a range of leukocytes to augment and bridge the innate and adaptive immune systems. Given the detrimental effect of chronic inflammation, the complement system is therefore well placed as an anti-inflammatory drug target. In this review, we provide a general outline of the sterile activators, effectors, and targets of the complement system and a series of examples (i.e., hypertension, cancer, allograft transplant rejection, and neuroinflammation) that highlight complement's ability to bridge the 2 arms of the immune system.
Collapse
Affiliation(s)
- Martin W Lo
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Benichou G, Wang M, Ahrens K, Madsen JC. Extracellular vesicles in allograft rejection and tolerance. Cell Immunol 2020; 349:104063. [PMID: 32087929 DOI: 10.1016/j.cellimm.2020.104063] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 01/19/2023]
Abstract
Extracellular vesicles (EVs), including exosomes, ectosomes and apoptotic vesicles, play an essential role in communication between cells of the innate and adaptive immune systems. Recent studies showed that EVs released after transplantation of allogeneic tissues and organs are involved in the immune recognition and response leading to rejection or tolerance in mice. After skin, pancreatic islet, and solid organ transplantation, donor-derived EVs were shown to initiate direct inflammatory alloresponses by T cells leading to acute rejection. This occurred through presentation of intact allogeneic MHC molecules on recipient antigen presenting cells (MHC cross-dressing) and subsequent activation of T cells via semi-direct allorecognition. On the other hand, some studies have documented the role of EVs in maternal tolerance of fetal alloantigens during pregnancy and immune privilege associated with spontaneous tolerance of liver allografts in laboratory rodents. The precise nature of the EVs, which are involved in rejection or tolerance, and the cells which produce them, is still unclear. Nevertheless, several reports showed that EVs released in the blood and urine by allografts can be used as biomarkers of rejection. This article reviews current knowledge on the contribution of EVs in allorecognition by T cells and discusses some mechanisms underlying their influence on T cell alloimmunity in allograft rejection or tolerance.
Collapse
Affiliation(s)
- Gilles Benichou
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| | - Mengchuan Wang
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Kaitlan Ahrens
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Joren C Madsen
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
13
|
Abstract
Despite decades of research, the induction and maintenance of long-term allograft tolerance without immunosuppression remains an elusive goal in the field of solid organ and cell transplantation. Immunosuppressive medications frequently prevent or minimize acute cellular rejection but have failed to halt antidonor antibody production and chronic organ rejection. Past efforts aimed at promoting lasting allograft tolerance have focused primarily on peripheral T-cell depletion, augmentation of regulatory T cells, or induction via simultaneous hematopoietic stem cell transplantation and facilitation of donor chimerism. So far, none of these methods have led to consistently safe, feasible and long lasting donor organ acceptance. Over the course of the past 4 decades, the study of a unique population of antigen-presenting cells known as dendritic cells has shown promise for breaking new ground in achieving indefinite allograft survival without immunosuppression and its associated adverse effects. In this review, we discuss the discovery and early investigations of dendritic cells and chronicle some of the key studies demonstrating their role in transplantation, particularly in indirect allorecognition, the immunologic pathway thought to drive chronic rejection and perhaps tolerance induction.
Collapse
|
14
|
Akbarpour M, Bharat A. Lung Injury and Loss of Regulatory T Cells Primes for Lung-Restricted Autoimmunity. Crit Rev Immunol 2019; 37:23-37. [PMID: 29431077 DOI: 10.1615/critrevimmunol.2017024944] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lung transplantation is a life-saving therapy for several end-stage lung diseases. However, lung allografts suffer from the lowest survival rate predominantly due to rejection. The pathogenesis of alloimmunity and its role in allograft rejection has been extensively studied and multiple approaches have been described to induce tolerance. However, in the context of lung transplantation, dysregulation of mechanisms, which maintain tolerance against self-antigens, can lead to lung-restricted autoimmunity, which has been recently identified to drive the immunopathogenesis of allograft rejection. Indeed, both preexisting as well as de novo lung-restricted autoimmunity can play a major role in the development of lung allograft rejection. The three most widely studied lung-restricted self-antigens include collagen type I, collagen type V, and k-alpha 1 tubulin. In this review, we discuss the role of lung-restricted autoimmunity in the development of both early as well as late lung allograft rejection and recent literature providing insight into the development of lung-restricted autoimmunity through the dysfunction of immune mechanisms which maintain peripheral tolerance.
Collapse
Affiliation(s)
- Mahzad Akbarpour
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ankit Bharat
- Division of Thoracic Surgery, Department of Surgery; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW This article reviews recent literature on the nature of extracellular vesicles released by allogeneic transplants and examine their role in T-cell alloimmunity involved in rejection and tolerance of these grafts. RECENT FINDINGS Donor cells release extracellular vesicles, including exosomes, after transplantation of allogeneic organs and tissues. Consequently, recipient APCs take up these exosomes and present donor MHC antigens on their surface (allo-MHC cross-dressing) thus, activating some alloreactive T cells via a mechanism called semi-direct pathway of allorecognition. In addition, one study shows that exosomes carrying noninherited maternal antigens are associated with maternal microchimerism and tolerance in offspring. Finally, a few studies describe potential utilization of exosomes as modulators of alloimmunity and biomarkers of rejection in allotransplantation. SUMMARY Extracellular vesicles, including exosomes, released by allografts contribute to recognition of donor antigens by T cells after allotransplantation. This occurs through cross-dressing of recipient APCs with donor MHC antigens and subsequent activation of T cells, a process called semi-direct alloreactivity. The relevance of this phenomenon in rejection and tolerance of allografts and the potential utilization of exosomes as biomarkers in transplantation are discussed.
Collapse
|
16
|
Hamers AAJ, Joshi SK, Pillai AB. Innate Immune Determinants of Graft-Versus-Host Disease and Bidirectional Immune Tolerance in Allogeneic Transplantation. ACTA ACUST UNITED AC 2019; 3. [PMID: 33511333 PMCID: PMC7839993 DOI: 10.21926/obm.transplant.1901044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The success of tissue transplantation from a healthy donor to a diseased individual (allo-transplantation) is regulated by the immune systems of both donor and recipient. Developing a state of specific non-reactivity between donor and recipient, while maintaining the salutary effects of immune function in the recipient, is called “immune (transplantation) tolerance”. In the classic early post-transplant period, minimizing bidirectional donor ←→ recipient reactivity requires the administration of immunosuppressive drugs, which have deleterious side effects (severe immunodeficiency, opportunistic infections, and neoplasia, in addition to drug-specific reactions and organ toxicities). Inducing immune tolerance directly through donor and recipient immune cells, particularly via subsets of immune regulatory cells, has helped to significantly reduce side effects associated with multiple immunosuppressive drugs after allo-transplantation. The innate and adaptive arms of the immune system are both implicated in inducing immune tolerance. In the present article, we will review innate immune subset manipulations and their potential applications in hematopoietic stem cell transplantation (HSCT) to cure malignant and non-malignant hematological disorders by inducing long-lasting donor ←→ recipient (bidirectional) immune tolerance and reduced graft-versus-host disease (GVHD). These innate immunotherapeutic strategies to promote long-term immune allo-transplant tolerance include myeloid-derived suppressor cells (MDSCs), regulatory macrophages, tolerogenic dendritic cells (tDCs), Natural Killer (NK) cells, invariant Natural Killer T (iNKT) cells, gamma delta T (γδ-T) cells and mesenchymal stromal cells (MSCs).
Collapse
Affiliation(s)
- Anouk A J Hamers
- Department of Pediatrics, Division of Hematology / Oncology and Bone Marrow Transplantation, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sunil K Joshi
- Department of Pediatrics, Division of Hematology / Oncology and Bone Marrow Transplantation, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Asha B Pillai
- Department of Pediatrics, Division of Hematology / Oncology and Bone Marrow Transplantation, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
17
|
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: 154] [Impact Index Per Article: 22.0] [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
|
18
|
Didona D, Di Zenzo G. Humoral Epitope Spreading in Autoimmune Bullous Diseases. Front Immunol 2018; 9:779. [PMID: 29719538 PMCID: PMC5913575 DOI: 10.3389/fimmu.2018.00779] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/28/2018] [Indexed: 12/15/2022] Open
Abstract
Autoimmune blistering diseases are characterized by autoantibodies against structural adhesion proteins of the skin and mucous membranes. Extensive characterization of their autoantibody targets has improved understanding of pathogenesis and laid the basis for the study of antigens/epitopes diversification, a process termed epitope spreading (ES). In this review, we have reported and discussed ES phenomena in autoimmune bullous diseases and underlined their functional role in disease pathogenesis. A functional ES has been proposed: (1) in bullous pemphigoid patients and correlates with the initial phase of the disease, (2) in pemphigus vulgaris patients with mucosal involvement during the clinical transition to a mucocutaneous form, (3) in endemic pemphigus foliaceus, underlining its role in disease pathogenesis, and (4) in numerous cases of disease transition associated with an intermolecular diversification of immune response. All these findings could give useful information to better understand autoimmune disease pathogenesis and to design antigen/epitope specific therapeutic approaches.
Collapse
Affiliation(s)
- Dario Didona
- Clinic for Dermatology and Allergology, University Hospital Marburg, University of Marburg, Marburg, Germany
| | - Giovanni Di Zenzo
- Molecular and Cell Biology Laboratory, Istituto Dermopatico dell’Immacolata (IDI)-IRCCS, Rome, Italy
| |
Collapse
|
19
|
Fejzić E, Karamehić J, Eminović I, Suljević D, Alijagić A, Bećirević S, Šahović A, Šišić S. HLA Genotyping in Patients with End-Stage Renal Disease Waiting For Cadaveric Renal Transplantation in Federation of Bosnia and Herzegovina. Open Access Maced J Med Sci 2017; 5:1-5. [PMID: 28293306 PMCID: PMC5320897 DOI: 10.3889/oamjms.2017.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/18/2016] [Accepted: 12/24/2016] [Indexed: 12/02/2022] Open
Abstract
AIM The research was conducted by genotyping two Human Leukocyte Antigen (HLA) gene classes. The main objective of this research was to investigate distribution and frequency of the allelic groups, genotypes and haplotypes in the gene loci of HLA class I (HLA-A*, -B*, -C*) and HLA class II (HLA-DRB1*, -DQB1*) in patients included in the program of cadaveric renal transplantation. MATERIAL AND METHODS Our study covered 186 blood samples of patients who are registered on the list for cadaveric renal transplantation in Federation of Bosnia and Herzegovina and included 59 control, healthy unrelated individuals. For the HLA typing, we have used three different methods: micro lymphocyte cytotoxicity test (MLCT), Polymerase Chain Reaction (PCR) - Sequence Specific Primers (SSP) and PCR - Sequence-Specific Oligonucleotides (SSO) or Luminex technology. All patients and cadaveric donors were tested using the three methods because the system is polymorphic. RESULTS Analysis of the results of genotyping HLA class I gene loci identified dominant HLA-A*02, HLA-B*35, HLA-C*07 allelic groups. Analysis of the HLA class II gene loci genotyping showed that HLA-DRB1*11 and HLA-DQB1*03 loci had the highest incidence in HLA class II. CONCLUSION Based on our results and previous research, there were no observed differences between allelic frequencies and genotypes of healthy people and people with ESRD. Differences between allelic groups occurred, but they were not statistically significant, except HLA-C*01 (p = 0.020).
Collapse
Affiliation(s)
- Elma Fejzić
- Institute of Transfusion Medicine of Federation of Bosnia and Herzegovina, Sarajevo, Bosnia and Herzegovina
| | - Jasenko Karamehić
- Department of Immunology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Izet Eminović
- Department of Biochemistry and Physiology, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Damir Suljević
- Department of Biochemistry and Physiology, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Andi Alijagić
- Department of Biochemistry and Physiology, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Semir Bećirević
- Institute of Pathology, Clinical Center University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Amela Šahović
- Institute of Transfusion Medicine of Federation of Bosnia and Herzegovina, Sarajevo, Bosnia and Herzegovina
| | - Sanela Šišić
- Institute of Transfusion Medicine of Federation of Bosnia and Herzegovina, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
20
|
da Silva MB, da Cunha FF, Terra FF, Camara NOS. Old game, new players: Linking classical theories to new trends in transplant immunology. World J Transplant 2017; 7:1-25. [PMID: 28280691 PMCID: PMC5324024 DOI: 10.5500/wjt.v7.i1.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/16/2016] [Accepted: 12/07/2016] [Indexed: 02/05/2023] Open
Abstract
The evolutionary emergence of an efficient immune system has a fundamental role in our survival against pathogenic attacks. Nevertheless, this same protective mechanism may also establish a negative consequence in the setting of disorders such as autoimmunity and transplant rejection. In light of the latter, although research has long uncovered main concepts of allogeneic recognition, immune rejection is still the main obstacle to long-term graft survival. Therefore, in order to define effective therapies that prolong graft viability, it is essential that we understand the underlying mediators and mechanisms that participate in transplant rejection. This multifaceted process is characterized by diverse cellular and humoral participants with innate and adaptive functions that can determine the type of rejection or promote graft acceptance. Although a number of mediators of graft recognition have been described in traditional immunology, recent studies indicate that defining rigid roles for certain immune cells and factors may be more complicated than originally conceived. Current research has also targeted specific cells and drugs that regulate immune activation and induce tolerance. This review will give a broad view of the most recent understanding of the allogeneic inflammatory/tolerogenic response and current insights into cellular and drug therapies that modulate immune activation that may prove to be useful in the induction of tolerance in the clinical setting.
Collapse
|
21
|
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
|
22
|
Graft dysfunction in chronic antibody-mediated rejection correlates with B-cell-dependent indirect antidonor alloresponses and autocrine regulation of interferon-γ production by Th1 cells. Kidney Int 2016; 91:477-492. [PMID: 27988211 PMCID: PMC5258815 DOI: 10.1016/j.kint.2016.10.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/19/2016] [Accepted: 10/06/2016] [Indexed: 12/22/2022]
Abstract
Chronic antibody-mediated rejection, a common cause of renal transplant failure, has a variable clinical phenotype. Understanding why some with chronic antibody-mediated rejection progress slowly may help develop more effective therapies. B lymphocytes act as antigen-presenting cells for in vitro indirect antidonor interferon-γ production in chronic antibody-mediated rejection, but many patients retain the ability to regulate these responses. Here we test whether particular patterns of T and B cell antidonor response associate with the variability of graft dysfunction in chronic antibody-mediated rejection. Our results confirm that dynamic changes in indirect antidonor CD4+ T-cell responses correlate with changes in estimated glomerular filtration rates, independent of other factors. Graft dysfunction progressed rapidly in patients who developed unregulated B-cell–driven interferon-γ production. However, conversion to a regulated or nonreactive pattern, which could be achieved by optimization of immunosuppression, associated with stabilization of graft function. Functional regulation by B cells appeared to activate an interleukin-10 autocrine pathway in CD4+ T cells that, in turn, impacted on antigen-specific responses. Thus, our data significantly enhance the understanding of graft dysfunction associated with chronic antibody-mediated rejection and provide the foundation for strategies to prolong renal allograft survival, based on regulation of interferon-γ production.
Collapse
|
23
|
Autoimmune Liver Disease Post-Liver Transplantation: A Summary and Proposed Areas for Future Research. Transplantation 2016; 100:515-24. [PMID: 26447505 PMCID: PMC4764021 DOI: 10.1097/tp.0000000000000922] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Autoimmune liver diseases (AILD) are rare diseases with a reported prevalence of less than 50 per 100 000 population. As the research landscape and our understanding of AILDs and liver transplantation evolves, there remain areas of unmet needs. One of these areas of unmet needs is prevention of disease recurrence after liver transplantation. Disease recurrence is not an insignificant event because allograft loss with the need for retransplantation can occur. Patients transplanted for AILD are more likely to experience acute rejection compared to those transplanted for non-AILD, and the reason(s) behind this observation is unclear. Tasks for the future include a better understanding of the pathogenesis of AILD, definition of the precise pathogenetic mechanisms of recurrent AILD, and development of strategies that can identify recipients at risk for disease recurrence. Importantly, the role of crosstalk between alloimmune responses and autoimmune responses in AILD is an important area that needs further study. This article reviews the relevant literature of de novo autoimmune hepatitis, recurrent autoimmune hepatitis, recurrent primary sclerosing cholangitis, and recurrent primary biliary cirrhosis in terms of the clinical entity, the scientific advancements, and future scientific goals to enhance our understanding of these diseases. A review of the relevant literature of de novo autoimmune hepatitis, recurrent autoimmune hepatitis, recurrent primary sclerosing cholangitis, and recurrent primary biliary cirrhosis in terms of the clinical entity, the scientific advancements and future scientific goals to enhance our understanding of these diseases.
Collapse
|
24
|
Bestard O, Cravedi P. Monitoring alloimmune response in kidney transplantation. J Nephrol 2016; 30:187-200. [PMID: 27245689 DOI: 10.1007/s40620-016-0320-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 05/15/2016] [Indexed: 01/22/2023]
Abstract
Currently, immunosuppressive therapy in kidney transplant recipients is generally performed by protocols and adjusted according to functional or histological evaluation of the allograft and/or signs of drug toxicity or infection. As a result, a large fraction of patients are likely to receive too much or too little immunosuppression, exposing them to higher rates of infection, malignancy and drug toxicity, or increased risk of acute and chronic graft injury from rejection, respectively. Developing reliable biomarkers is crucial for individualizing therapy aimed at extending allograft survival. Emerging data indicate that many assays, likely used in panels rather than single assays, have potential to be diagnostic and predictive of short and also long-term outcome. While numerous cross-sectional studies have found associations between the results of these assays and the presence of clinically relevant post-transplantation outcomes, data from prospective studies are still scanty, thereby preventing widespread implementation in the clinic. Of note, some prospective, randomized, multicenter biomarker-driven studies are currently on-going aiming at confirming such preliminary data. These works as well as other future studies are highly warranted to test the hypothesis that tailoring immunosuppression on the basis of results offered by these biomarkers leads to better outcomes than current standard clinical practice.
Collapse
Affiliation(s)
- Oriol Bestard
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona University, IDIBELL, Barcelona, Spain
| | - Paolo Cravedi
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Annenberg Building, New York, NY, 10029, USA.
| |
Collapse
|
25
|
Ali JM, Negus MC, Conlon TM, Harper IG, Qureshi MS, Motallebzadeh R, Willis R, Saeb-Parsy K, Bolton EM, Bradley JA, Pettigrew GJ. Diversity of the CD4 T Cell Alloresponse: The Short and the Long of It. Cell Rep 2016; 14:1232-1245. [PMID: 26804905 PMCID: PMC5405053 DOI: 10.1016/j.celrep.2015.12.099] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 11/23/2015] [Accepted: 12/21/2015] [Indexed: 01/03/2023] Open
Abstract
MHC alloantigen is recognized by two pathways: "directly," intact on donor cells, or "indirectly," as self-restricted allopeptide. The duration of each pathway, and its relative contribution to allograft vasculopathy, remain unclear. Using a murine model of chronic allograft rejection, we report that direct-pathway CD4 T cell alloresponses, as well as indirect-pathway responses against MHC class II alloantigen, are curtailed by rapid elimination of donor hematopoietic antigen-presenting cells. In contrast, persistent presentation of epitope resulted in continual division and less-profound contraction of the class I allopeptide-specific CD4 T cell population, with approximately 10,000-fold more cells persisting than following acute allograft rejection. This expanded population nevertheless displayed sub-optimal anamnestic responses and was unable to provide co-stimulation-independent help for generating alloantibody. Indirect-pathway CD4 T cell responses are heterogeneous. Appreciation that responses against particular alloantigens dominate at late time points will likely inform development of strategies aimed at improving transplant outcomes.
Collapse
Affiliation(s)
- Jason M Ali
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Margaret C Negus
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Thomas M Conlon
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Ines G Harper
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - M Saeed Qureshi
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Reza Motallebzadeh
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Richard Willis
- NIH Tetramer Facility, Emory/Yerkes, 954 Gatewood Road, Atlanta, GA 30329, USA
| | - Kourosh Saeb-Parsy
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Eleanor M Bolton
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - J Andrew Bradley
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Gavin J Pettigrew
- University of Cambridge, School of Clinical Medicine, Cambridge CB2 0QQ, UK.
| |
Collapse
|
26
|
Starling RC, Stehlik J, Baran DA, Armstrong B, Stone JR, Ikle D, Morrison Y, Bridges ND, Putheti P, Strom TB, Bhasin M, Guleria I, Chandraker A, Sayegh M, Daly KP, Briscoe DM, Heeger PS. Multicenter Analysis of Immune Biomarkers and Heart Transplant Outcomes: Results of the Clinical Trials in Organ Transplantation-05 Study. Am J Transplant 2016; 16:121-36. [PMID: 26260101 PMCID: PMC4948061 DOI: 10.1111/ajt.13422] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/20/2015] [Accepted: 06/14/2015] [Indexed: 01/25/2023]
Abstract
Identification of biomarkers that assess posttransplant risk is needed to improve long-term outcomes following heart transplantation. The Clinical Trials in Organ Transplantation (CTOT)-05 protocol was an observational, multicenter, cohort study of 200 heart transplant recipients followed for the first posttransplant year. The primary endpoint was a composite of death, graft loss/retransplantation, biopsy-proven acute rejection (BPAR), and cardiac allograft vasculopathy (CAV) as defined by intravascular ultrasound (IVUS). We serially measured anti-HLA- and auto-antibodies, angiogenic proteins, peripheral blood allo-reactivity, and peripheral blood gene expression patterns. We correlated assay results and clinical characteristics with the composite endpoint and its components. The composite endpoint was associated with older donor allografts (p < 0.03) and with recipient anti-HLA antibody (p < 0.04). Recipient CMV-negativity (regardless of donor status) was associated with BPAR (p < 0.001), and increases in plasma vascular endothelial growth factor-C (OR 20; 95%CI:1.9-218) combined with decreases in endothelin-1 (OR 0.14; 95%CI:0.02-0.97) associated with CAV. The remaining biomarkers showed no relationships with the study endpoints. While suboptimal endpoint definitions and lower than anticipated event rates were identified as potential study limitations, the results of this multicenter study do not yet support routine use of the selected assays as noninvasive approaches to detect BPAR and/or CAV following heart transplantation.
Collapse
Affiliation(s)
| | - Josef Stehlik
- University of Utah Health Sciences Center, Salt Lake City UT
| | | | | | | | | | - Yvonne Morrison
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD
| | - Nancy D. Bridges
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Bhatti AB, Usman M. Chronic Renal Transplant Rejection and Possible Anti-Proliferative Drug Targets. Cureus 2015; 7:e376. [PMID: 26677426 PMCID: PMC4671911 DOI: 10.7759/cureus.376] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/06/2015] [Indexed: 12/17/2022] Open
Abstract
The global prevalence of renal transplants is increasing with time, and renal transplantation is the only definite treatment for end-stage renal disease. We have limited the acute and late acute rejection of kidney allografts, but the long-term survival of renal tissues still remains a difficult and unanswered question as most of the renal transplants undergo failure within a decade of their transplantation. Among various histopathological changes that signify chronic allograft nephropathy (CAN), tubular atrophy, fibrous thickening of the arteries, fibrosis of the kidney interstitium, and glomerulosclerosis are the most important. Moreover, these structural changes are followed by a decline in the kidney function as well. The underlying mechanism that triggers the long-term rejection of renal transplants involves both humoral and cell-mediated immunity. T cells, with their related cytokines, cause tissue damage. In addition, CD 20+ B cells and their antibodies play an important role in the long-term graft rejection. Other risk factors that predispose a recipient to long-term graft rejection include HLA-mismatching, acute episodes of graft rejection, mismatch in donor-recipient age, and smoking. The purpose of this review article is the analyze current literature and find different anti-proliferative agents that can suppress the immune system and can thus contribute to the long-term survival of renal transplants. The findings of this review paper can be helpful in understanding the long-term survival of renal transplants and various ways to improve it.
Collapse
Affiliation(s)
- Adnan Bashir Bhatti
- Department of Medicine, Capital Development Authority Hospital, Islamabad, Pakistan
| | - Muhammad Usman
- Department of Medicine, Jinnah Hospital Lahore (JHL)/Allama Iqbal Medical College (AIMC), Lahore, Pakistan
| |
Collapse
|
28
|
Abstract
Each individual harbours a unique set of commensal microorganisms, collectively referred to as the microbiota. Notably, these microorganisms exceed the number of cells in the human body by 10-fold. This finding has accelerated a shift in our understanding of human physiology, with the realization that traits necessary for health are both encoded and influenced by the human genome and the microbiota. Our understanding of the aetiology of complex diseases has, therefore, evolved with increasing awareness that the human microbiota has an active and critical role in maintaining health and inducing disease. Indeed, findings from bioinformatic studies indicate that the microbiota and microbiome have multiple effects on the innate and adaptive immune systems, with effects on infection, autoimmune disease and cancer. In this Review, we first address the important statistical and informatics aspects that should be considered when characterizing the composition of microbiota. We next highlight the effects of the microbiota on the immune system and the implications of these effects on organ failure and transplantation. Finally, we reflect on the future perspectives for studies of the microbiota, including novel diagnostic tests and therapeutics.
Collapse
|
29
|
Mehrotra A, Leventhal J, Purroy C, Cravedi P. Monitoring T cell alloreactivity. Transplant Rev (Orlando) 2014; 29:53-9. [PMID: 25475045 DOI: 10.1016/j.trre.2014.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/03/2014] [Accepted: 11/09/2014] [Indexed: 01/06/2023]
Abstract
Currently, immunosuppressive therapy in kidney transplant recipients is center-specific, protocol-driven, and adjusted according to functional or histological evaluation of the allograft and/or signs of drug toxicity or infection. As a result, a large fraction of patients receive too much or too little immunosuppression, exposing them to higher rates of infection, malignancy and drug toxicity, or increased risk of acute and chronic graft injury from rejection, respectively. The individualization of immunosuppression requires the development of assays able to reliably quantify and/or predict the magnitude of the recipient's immune response toward the allograft. As alloreactive T cells are central mediators of allograft rejection, monitoring T cell alloreactivity has become a priority for the transplant community. Among available assays, flow cytometry based phenotyping, T cell proliferation, T cell cytokine secretion, and ATP release (ImmuKnow), have been the most thoroughly tested. While numerous cross-sectional studies have found associations between the results of these assays and the presence of clinically relevant post-transplantation outcomes, data from prospective studies are still scanty, thereby preventing widespread implementation in the clinic. Future studies are required to test the hypothesis that tailoring immunosuppression on the basis of results offered by these biomarkers leads to better outcomes than current standard clinical practice.
Collapse
Affiliation(s)
- Anita Mehrotra
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Jeremy Leventhal
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Carolina Purroy
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Paolo Cravedi
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, USA.
| |
Collapse
|
30
|
Thus KA, Ruizendaal MT, de Hoop TA, Borst E, van Deutekom HW, te Boome L, Kuball J, Spierings E. Refinement of the Definition of Permissible HLA-DPB1 Mismatches with Predicted Indirectly ReCognizable HLA-DPB1 Epitopes. Biol Blood Marrow Transplant 2014; 20:1705-10. [DOI: 10.1016/j.bbmt.2014.06.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/19/2014] [Indexed: 01/08/2023]
|
31
|
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
|
32
|
Mierzejewska B, Schroder PM, Baum CE, Blair A, Smith C, Duquesnoy RJ, Marrari M, Gohara A, Malhotra D, Kaw D, Liwski R, Rees MA, Stepkowski S. Early acute antibody-mediated rejection of a negative flow crossmatch 3rd kidney transplant with exclusive disparity at HLA-DP. Hum Immunol 2014; 75:703-8. [PMID: 24755353 DOI: 10.1016/j.humimm.2014.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 02/27/2014] [Accepted: 04/05/2014] [Indexed: 02/02/2023]
Abstract
Donor-specific alloantibodies (DSA) to HLA-DP may cause antibody-mediated rejection (AMR), especially in re-transplants. We describe the immunization history of a patient who received 3 kidney transplants; the 3rd kidney was completely matched except at DPA1 and DPB1. Prior to the 3rd transplant, single antigen bead analysis (SAB) showed DSA reactivity against DPA1 shared by the 1st and 3rd donors, but B and T flow crossmatch (FXM) results were negative. Within 11 days the 3rd transplant underwent acute C4d+ AMR which coincided with the presence of complement (C1q)-binding IgG1 DSA against donor DPA1 and DPB1. Using HLAMatchmaker and SAB, we provide evidence that eplet (epitope) spreading on DPA1 and eplet sharing on differing DPB1 alleles of the 1st and 3rd transplants was associated with AMR. Since weak DSA to DPA1/DPB1 may induce acute AMR with negative FXM, donor DPA1/DPB1 high resolution typing should be considered in sensitized patients with DP-directed DSA.
Collapse
Affiliation(s)
- Beata Mierzejewska
- Department of Urology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Paul M Schroder
- Department of Microbiology and Immunology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Caitlin E Baum
- Department of Microbiology and Immunology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Annette Blair
- Department of Pathology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Connie Smith
- Department of Pathology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Rene J Duquesnoy
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Marilyn Marrari
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Amira Gohara
- Department of Pathology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Deepak Malhotra
- Department of Internal Medicine, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Dinkar Kaw
- Department of Internal Medicine, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Robert Liwski
- HLA Typing Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael A Rees
- Department of Urology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Stanislaw Stepkowski
- Department of Microbiology and Immunology, University of Toledo College of Medicine, Toledo, OH 43614, USA.
| |
Collapse
|
33
|
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]
|
34
|
Tolerogenic dendritic cells and induction of T suppressor cells in transplant recipients. Methods Mol Biol 2014; 1034:359-71. [PMID: 23775751 DOI: 10.1007/978-1-62703-493-7_23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Tolerogenic antigen presenting cells (APC), primarily dendritic cells (DC), are essential to the induction and maintenance of immunologic tolerance in clinical transplantation. They induce the differentiation of CD8+ T suppressor (Ts) and CD4+ T regulatory (Treg) or anergic cells, which prevent transplant rejection maintaining a state of quiescence. Tolerogenic APC express high levels of inhibitory receptors such as Immunoglobulin-like transcript (ILT)3 and 4 which inhibit the effector function of T cells that recognize HLA-peptide complexes on APC. Here, we describe the methods for detection of tolerogenic APC induced by allospecific Ts/Treg cells.
Collapse
|
35
|
Ballet C, Giral M, Ashton-Chess J, Renaudin K, Brouard S, Soulillou JP. Chronic rejection of human kidney allografts. Expert Rev Clin Immunol 2014; 2:393-402. [DOI: 10.1586/1744666x.2.3.393] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
36
|
Abstract
With the advent of cellular therapies, it has become clear that the success of future therapies in prolonging allograft survival will require an intimate understanding of the allorecognition pathways and effector mechanisms that are responsible for chronic rejection and late graft loss.Here, we consider current understanding of T-cell allorecognition pathways and discuss the most likely mechanisms by which these pathways collaborate with other effector mechanisms to cause allograft rejection. We also consider how this knowledge may inform development of future strategies to prevent allograft rejection.Although both direct and indirect pathway CD4 T cells appear active immediately after transplantation, it has emerged that indirect pathway CD4 T cells are likely to be the dominant alloreactive T-cell population late after transplantation. Their ability to provide help for generating long-lived alloantibody is likely one of the main mechanisms responsible for the progression of allograft vasculopathy and chronic rejection.Recent work has suggested that regulatory T cells may be an effective cellular therapy in transplantation. Given the above, adoptive therapy with CD4 regulatory T cells with indirect allospecificity is a rational first choice in attempting to attenuate the development and progression of chronic rejection; those with additional properties that enable inhibition of germinal center alloantibody responses hold particular appeal.
Collapse
|
37
|
Low Serum Testosterone as a New Risk Factor for Chronic Rejection in Heart Transplanted Men. Transplantation 2013; 96:501-5. [DOI: 10.1097/tp.0b013e31829b0893] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Ichikawa K, Kagamu H, Koyama K, Miyabayashi T, Koshio J, Miura S, Watanabe S, Yoshizawa H, Narita I. Epitope diversification driven by non-tumor epitope-specific Th1 and Th17 mediates potent antitumor reactivity. Vaccine 2012; 30:6190-7. [PMID: 22889826 DOI: 10.1016/j.vaccine.2012.07.060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 07/03/2012] [Accepted: 07/24/2012] [Indexed: 10/28/2022]
Abstract
MHC class I-restricted peptide-based vaccination therapies have been conducted to treat cancer patients, because CD8⁺ CTL can efficiently induce apoptosis of tumor cells in an MHC class I-restricted epitope-specific manner. Interestingly, clinical responders are known to demonstrate reactivity to epitopes other than those used for vaccination; however, the mechanism underlying how antitumor T cells with diverse specificity are induced is unclear. In this study, we demonstrated that dendritic cells (DCs) that engulfed apoptotic tumor cells in the presence of non-tumor MHC class II-restricted epitope peptides, OVA(323-339), efficiently presented tumor-associated antigens upon effector-dominant CD4⁺ T cell balance against regulatory T cells (Treg) for the OVA(323-339) epitope. Th1 and Th17 induced tumor-associated antigens presentation of DC, while Th2 ameliorated tumor-antigen presentation for CD8⁺ T cells. Blocking experiments with anti-IL-23p19 antibody and anti-IL-23 receptor indicated that an autocrine mechanism of IL-23 likely mediated the diverted tumor-associated antigens presentation of DC. Tumor-associated antigens presentation of DC induced by OVA(323-339) epitope-specific CD4⁺ T cells resulted in facilitated antitumor immunity in both priming and effector phase in vivo. Notably, this immunotherapy did not require pretreatment to reduce Treg induced by tumor. This strategy may have clinical implications for designing effective antitumor immunotherapies.
Collapse
Affiliation(s)
- Kosuke Ichikawa
- Division of Respiratory Medicine, Department of Homeostatic Regulation and Development, Course for Biological Functions and Medical Control, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sagoo P, Lombardi G, Lechler RI. Relevance of regulatory T cell promotion of donor-specific tolerance in solid organ transplantation. Front Immunol 2012; 3:184. [PMID: 22811678 PMCID: PMC3395995 DOI: 10.3389/fimmu.2012.00184] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 06/14/2012] [Indexed: 01/29/2023] Open
Abstract
Current clinical strategies to control the alloimmune response after transplantation do not fully prevent induction of the immunological processes which lead to acute and chronic immune-mediated graft rejection, and as such the survival of a solid organ allograft is limited. Experimental research on naturally occurring CD4+CD25highFoxP3+ Regulatory T cells (Tregs) has indicated their potential to establish stable long-term graft acceptance, with the promise of providing a more effective therapy for transplant recipients. Current approaches for clinical use are based on the infusion of freshly isolated or ex vivo polyclonally expanded Tregs into graft recipients with an aim to redress the in vivo balance of T effector cells to Tregs. However mounting evidence suggests that regulation of donor-specific immunity may be central to achieving immunological tolerance. Therefore, the next stages in optimizing translation of Tregs to organ transplantation will be through the refinement and development of donor alloantigen-specific Treg therapy. The altering kinetics and intensity of alloantigen presentation pathways and alloimmune priming following transplantation may indeed influence the specificity of the Treg required and the timing or frequency at which it needs to be administered. Here we review and discuss the relevance of antigen-specific regulation of alloreactivity by Tregs in experimental and clinical studies of tolerance and explore the concept of delivering an optimal Treg for the induction and maintenance phases of achieving transplantation tolerance.
Collapse
Affiliation(s)
- Pervinder Sagoo
- Department Transplantation, Immunoregulation and Mucosal Biology, MRC Centre for Transplantation, King's College London London, UK
| | | | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Chronic injury and late allograft loss remain major causes of morbidity in clinical transplantation. Biomarkers that can reliably assess the risk of posttransplant complications are required to direct and individualize therapy aimed at prolonging graft survival and improving patient health. The purpose of this review is to provide a framework for understanding how to use biomarkers in the context of clinical transplantation and to summarize current data on available noninvasive cellular-based immune monitoring methods to predict transplant outcome. RECENT FINDINGS New microarray and gene profiling data reveal peripheral blood cell gene expression patterns that identify operational tolerance, raising the possibility that the measurements can be used to direct immunosuppression withdrawal. Additional data support the use of selective urine gene products and soluble CD30 measurements in serum as reliable biomarkers of acute graft injury. Finally, recent studies demonstrate that measurement of T-cell alloimmunity by cytokine enzyme-linked immunospot is a promising, supplementary pretransplant risk assessment tool. SUMMARY Recently published studies in organ transplantation suggest that results derived from assays focused on markers of T-cell immunity can segregate transplant candidates or recipients into high and low-risk subgroups for posttransplant graft injury. Larger prospective studies are needed, however, before any proposed biomarker can be incorporated into the transplant physicians' armamentarium to guide individualized therapeutic decision-making.
Collapse
|
41
|
Are we ready for the use of foxp3(+) regulatory T cells for immunodiagnosis and immunotherapy in kidney transplantation? J Transplant 2012; 2012:397952. [PMID: 22690325 PMCID: PMC3368592 DOI: 10.1155/2012/397952] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/19/2012] [Indexed: 02/06/2023] Open
Abstract
The existence of T-cell subsets naturally committed to perform immunoregulation has led to enthusiastic efforts to investigate their role in the immunopathogenesis of transplantation. Being able to modulate alloresponses, regulatory T cells could be used as an immunodiagnostic tool in clinical kidney transplantation. Thus, the measurement of Foxp3 transcripts, the presence of regulatory T cells in kidney biopsies, and the phenotypic characterisation of the T-cell infiltrate could aid in the diagnosis of rejection and the immune monitoring and prediction of outcomes in kidney transplantation. Interestingly, the adoptive transfer of regulatory T cells in animal models has been proven to downmodulate powerful alloresponses, igniting translational research on their potential use as an immunomodulatory therapy. For busy transplant clinicians, the vast amount of information in the literature on regulatory T cells can be overwhelming. This paper aims to highlight the most applicable research findings on the use of regulatory T cells in the immune diagnosis and potential immunomodulatory therapy of kidney transplant patients. However, can we yet rely on differential regulatory T-cell profiles for the identification of rejection or to tailor patient's immunosuppression? Are we ready to administer regulatory T cells as inductive or adjunctive therapy for kidney transplantation?
Collapse
|
42
|
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
|
43
|
Tiriveedhi V, Angaswamy N, Brand D, Weber J, Gelman AG, Hachem R, Trulock EP, Meyers B, Patterson G, Mohanakumar T. A shift in the collagen V antigenic epitope leads to T helper phenotype switch and immune response to self-antigen leading to chronic lung allograft rejection. Clin Exp Immunol 2012; 167:158-68. [PMID: 22132895 DOI: 10.1111/j.1365-2249.2011.04486.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Immune responses to human leucocyte antigen (HLA) and self-antigen collagen V (Col-V) have been proposed in the pathogenesis of chronic rejection (bronchiolitis obliterans syndrome, BOS) following human lung transplantation (LTx). In this study, we defined the role for the shift in immunodominant epitopes of Col-V in inducing T helper phenotype switch leading to immunity to Col-V and BOS. Sera and lavage from BOS(+) LTx recipients with antibodies to Col-V were analysed. Two years prior to BOS, patients developed antibodies to both Col-V,α1(V) and α2(V) chains. However, at clinical diagnosis of BOS, antibodies became restricted to α1(V). Further, lung biopsy from BOS(+) patients bound to antibodies to α1(V), indicating that these epitopes are exposed. Fourteen Col-V peptides [pep1-14, pep1-4 specific to α1(V), pep5-8 to α1,2(V) and pep9-14 to α2(V)] which bind to HLA-DR4 and -DR7, demonstrated that prior to BOS, pep 6, 7, 9, 11 and 14 were immunodominant and induced interleukin (IL)-10. However, at BOS, the response switched to pep1, 4 and 5 and induced interferon (IFN)-γ and IL-17 responses, but not IL-10. The T helper (Th) phenotype switch is accompanied by decreased frequency of regulatory T cells (T(regs) ) in the lavage. LTx recipients with antibodies to α1(V) also demonstrated increased matrix metalloproteinase (MMP) activation with decreased MMP inhibitor, tissue inhibitor of metalloproteinase (TIMP), suggesting that MMP activation may play a role in the exposure of new Col-V antigenic epitopes. We conclude that a shift in immunodominance of self-antigenic determinants of Col-V results in induction of IFN-γ and IL-17 with loss of tolerance leading to autoimmunity to Col-V, which leads to chronic lung allograft rejection.
Collapse
Affiliation(s)
- V Tiriveedhi
- Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Smith C, Miles JJ, Khanna R. Advances in direct T-cell alloreactivity: function, avidity, biophysics and structure. Am J Transplant 2012; 12:15-26. [PMID: 22152064 DOI: 10.1111/j.1600-6143.2011.03863.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Although T-cell-based adaptive immunity plays a crucial role in protection against infectious pathogens and uncontrolled outgrowth of malignant cells, a large portion of these T cells are also capable of responding to allogeneic HLA molecules, violating the paradigm of self-major histocompatibility complex (MHC) restriction. Recent studies have provided insights into the mechanisms by which these T cells recognize allogeneic targets. The role of antiviral T cells in direct alloreactivity through peptide-dependent molecular mimicry and alternate peptide-MHC docking modes has emerged as major models for the human alloresponse. Here, we review in depth recent advances in this field and discuss how molecular interactions between T cells and HLA molecules drive the activation of these effector cells and its potential implications for alloreactivity in human transplantation.
Collapse
Affiliation(s)
- C Smith
- Australian Centre for Vaccine Development, Tumour Immunology Laboratory, Queensland Institute of Medical Research, Herston, Brisbane, Australia
| | | | | |
Collapse
|
45
|
Nadazdin O, Boskovic S, Wee SL, Sogawa H, Koyama I, Colvin RB, Smith RN, Tocco G, O'Connor DH, Karl JA, Madsen JC, Sachs DH, Kawai T, Cosimi AB, Benichou G. Contributions of direct and indirect alloresponses to chronic rejection of kidney allografts in nonhuman primates. THE JOURNAL OF IMMUNOLOGY 2011; 187:4589-97. [PMID: 21957140 DOI: 10.4049/jimmunol.1003253] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The relative contribution of direct and indirect allorecognition pathways to chronic rejection of allogeneic organ transplants in primates remains unclear. In this study, we evaluated T and B cell alloresponses in cynomolgus monkeys that had received combined kidney/bone marrow allografts and myeloablative immunosuppressive treatments. We measured donor-specific direct and indirect T cell responses and alloantibody production in monkeys (n = 5) that did not reject their transplant acutely but developed chronic humoral rejection (CHR) and in tolerant recipients (n = 4) that never displayed signs of CHR. All CHR recipients exhibited high levels of anti-donor Abs and mounted potent direct T cell alloresponses in vitro. Such direct alloreactivity could be detected for more than 1 y after transplantation. In contrast, only two of five monkeys with CHR had a detectable indirect alloresponse. No indirect alloresponse by T cells and no alloantibody responses were found in any of the tolerant monkeys. Only one of four tolerant monkeys displayed a direct T cell alloresponse. These observations indicate that direct T cell alloresponses can be sustained for prolonged periods posttransplantation and result in alloantibody production and chronic rejection of kidney transplants, even in the absence of detectable indirect alloreactivity.
Collapse
Affiliation(s)
- Ognjenka Nadazdin
- Department of Surgery, Transplant Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Colvin-Adams M, Agnihotri A. Cardiac allograft vasculopathy: current knowledge and future direction. Clin Transplant 2011; 25:175-84. [PMID: 21457328 DOI: 10.1111/j.1399-0012.2010.01307.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cardiac allograft vasculopathy (CAV) is a unique form of coronary artery disease affecting heart transplant recipients. Although prognosis of heart transplant recipients has improved over time, CAV remains a significant cause of mortality beyond the first year of cardiac transplantation. Many traditional and non-traditional risk factors for the development of CAV have been described. Traditional risk factors include dyslipidemia, diabetes and hypertension. Non-traditional risk factors include cytomegalovirus infection, HLA mismatch, antibody-mediated rejection, and mode of donor brain death. There is a complex interplay between immunological and non-immunological factors ultimately leading to endothelial injury and exaggerated repair response. Pathologically, CAV manifests as fibroelastic proliferation of intima and luminal stenosis. Early diagnosis is paramount as heart transplant recipients are frequently asymptomatic owing to cardiac denervation related to the transplant surgery. Intravascular ultrasound (IVUS) offers many advantages over conventional angiography and is an excellent predictor of prognosis in heart transplant recipients. Many non-invasive diagnostic tests including dobutamine stress echocardiography, CT angiography, and MRI are available; though, none has replaced angiography. This review discusses the risk factors, pathogenesis, and diagnosis of CAV and highlights some current concepts and recent developments in this field.
Collapse
|
47
|
Ho EK, Vasilescu ER, Vlad G, Marboe CC, Addonizio LJ, Suciu-Foca N. HLA antibodies in pediatric heart transplantation. Pediatr Transplant 2011; 15:458-64. [PMID: 21450008 DOI: 10.1111/j.1399-3046.2010.01435.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We have analyzed the impact of anti-HLA antibodies present in the patients' circulation prior and/or following heart transplantation in a population of 108 pediatric recipients. Anti-HLA class I and class II antibodies were monitored by traditional CDC using donor and panel T and B lymphocytes and by SPA for detection of DSA. There was a highly significant correlation between the development of AMR and presence of CDC- or SPA-detected DSA. However, the fraction of the transplant population which remained AMR-free was much higher among patients with SPA-detected compared to CDC-detected DSA. Furthermore, long-term graft survival was negatively affected only by cytotoxic, complement-fixing anti-HLA class I antibodies developing following transplantation. Anti-HLA class I or class II antibodies detected by SPA had no effect on long-term survival rates.
Collapse
Affiliation(s)
- Eric K Ho
- Departments of Pathology & Cell Biology Pediatrics, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
48
|
Kalache S, Dinavahi R, Pinney S, Mehrotra A, Cunningham MW, Heeger PS. Anticardiac myosin immunity and chronic allograft vasculopathy in heart transplant recipients. THE JOURNAL OF IMMUNOLOGY 2011; 187:1023-30. [PMID: 21677143 DOI: 10.4049/jimmunol.1004195] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chronic allograft vasculopathy (CAV) contributes to heart transplant failure, yet its pathogenesis is incompletely understood. Although cellular and humoral alloimmunity are accepted pathogenic mediators, animal models suggest that T cells and Abs reactive to graft-expressed autoantigens, including cardiac myosin (CM), could participate. To test the relationship between CAV and anti-CM autoimmunity in humans, we performed a cross-sectional study of 72 heart transplant recipients: 40 with CAV and 32 without. Sera from 65% of patients with CAV contained anti-CM Abs, whereas <10% contained Abs to other autoantigens (p < 0.05), and only 18% contained anti-HLA Abs (p < 0.05 versus anti-CM). In contrast, 13% of sera from patients without CAV contained anti-CM Abs (p < 0.05; odds ratio [OR], associating CAV with anti-CM Ab = 13, 95% confidence interval [CI] 3.79-44.6). Multivariable analysis confirmed the association to be independent of time posttransplant and the presence of anti-HLA Abs (OR = 28, 95% CI 5.77-133.56). PBMCs from patients with CAV responded more frequently to, and to a broader array of, CM-derived peptides than those without CAV (p = 0.01). Detection of either CM-peptide-reactive T cells or anti-CM Abs was highly and independently indicative of CAV (OR = 45, 95% CI 4.04-500.69). Our data suggest detection of anti-CM immunity could be used as a biomarker for outcome in heart transplantation recipients and support the need for further studies to assess whether anti-CM immunity is a pathogenic mediator of CAV.
Collapse
Affiliation(s)
- Safa Kalache
- Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
49
|
Dinavahi R, George A, Tretin A, Akalin E, Ames S, Bromberg JS, Deboccardo G, Dipaola N, Lerner SM, Mehrotra A, Murphy BT, Nadasdy T, Paz-Artal E, Salomon DR, Schröppel B, Sehgal V, Sachidanandam R, Heeger PS. Antibodies reactive to non-HLA antigens in transplant glomerulopathy. J Am Soc Nephrol 2011; 22:1168-78. [PMID: 21566057 DOI: 10.1681/asn.2010111183] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Although T and B cell alloimmunity contribute to transplant injury, autoimmunity directed at kidney-expressed, non-HLA antigens may also participate. Because the specificity, prevalence, and importance of antibodies to non-HLA antigens in late allograft injury are poorly characterized, we used a protein microarray to compare antibody repertoires in pre- and post-transplant sera from several cohorts of patients with and without transplant glomerulopathy. Transplantation routinely induced changes in antibody repertoires, but we did not identify any de novo non-HLA antibodies common to patients with transplant glomerulopathy. The screening studies identified three reactivities present before transplantation that persisted after transplant and strongly associated with transplant glomerulopathy. ELISA confirmed that reactivity against peroxisomal-trans-2-enoyl-coA-reductase strongly associated with the development of transplant glomerulopathy in independent validation sets. In addition to providing insight into effects of transplantation on non-HLA antibody repertoires, these results suggest that pretransplant serum antibodies to peroxisomal-trans-2-enoyl-coA-reductase may predict prognosis in kidney transplantation.
Collapse
Affiliation(s)
- Rajani Dinavahi
- Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
T Lymphocyte Responses to Nonpolymorphic HLA-Derived Peptides Are Associated With Chronic Renal Allograft Dysfunction. Transplantation 2011; 91:279-86. [DOI: 10.1097/tp.0b013e318203862d] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|