1
|
O'Hara PE, Gorrai A, Farr M, Peltz M, Beaini H, Moayedi Y, Chih S, Truby LK. Revisiting Biomarkers of Cardiac Allograft Vasculopathy: Addressing the Achilles Heel of Heart Transplantation. Curr Heart Fail Rep 2024; 21:580-590. [PMID: 39414739 DOI: 10.1007/s11897-024-00685-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/18/2024]
Abstract
Nearly half of heart transplant recipients will be diagnosed with cardiac allograft vasculopathy (CAV) within five years after transplantation. Advanced CAV can lead to worsening heart failure as well as arrhythmias and sudden cardiac death. The only curative therapy for end-stage CAV is re-transplantation. Current diagnostic methods are invasive and limited by poor sensitivity in early disease. Despite its high prevalence in the post-transplantpopulation, the underlying pathophysiology of this condition has yet to be fully described. It is thought to be primarily related to endothelial dysfunction, immune activation, and cardiometabolic disease. Biomarkers reflecting these underlying processes, particularly endothelial injury and immune activation, have shown early promise in discriminating prevalent CAV. Next-generation sequencing technologies such as proteomic and transcriptomic profiling have also provided further insight into the pathophysiology of CAV through the identification of novel biomarkers. Ultimately, these biomarkers may have a role in not only diagnosing CAV but also highlighting potential targets for disease-specific therapies. In this article, we review the current data for biomarkers in CAV and discuss future directions for biomarker identification..
Collapse
Affiliation(s)
- Patrick E O'Hara
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ananya Gorrai
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maryjane Farr
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthias Peltz
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hadi Beaini
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Sharon Chih
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Lauren K Truby
- University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Advanced Heart Failure and Transplantation, Department of Medicine, Division of Cardiology, UT Southwestern Medical Center, 5959 Harry Hines Boulevard, Dallas, TX, 75390, USA.
| |
Collapse
|
2
|
Pober JS, Chih S, Kobashigawa J, Madsen JC, Tellides G. Cardiac allograft vasculopathy: current review and future research directions. Cardiovasc Res 2021; 117:2624-2638. [PMID: 34343276 PMCID: PMC8783389 DOI: 10.1093/cvr/cvab259] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/02/2021] [Accepted: 07/29/2021] [Indexed: 12/25/2022] Open
Abstract
Cardiac allograft vasculopathy (CAV) is a pathologic immune-mediated remodelling of the vasculature in transplanted hearts and, by impairing perfusion, is the major cause of late graft loss. Although best understood following cardiac transplantation, similar forms of allograft vasculopathy occur in other vascularized organ grafts and some features of CAV may be shared with other immune-mediated vasculopathies. Here, we describe the incidence and diagnosis, the nature of the vascular remodelling, immune and non-immune contributions to pathogenesis, current therapies, and future areas of research in CAV.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Coronary Artery Disease/epidemiology
- Coronary Artery Disease/immunology
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/pathology
- Coronary Vessels/immunology
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Endothelial Cells/immunology
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Graft Rejection/epidemiology
- Graft Rejection/immunology
- Graft Rejection/metabolism
- Graft Rejection/pathology
- Graft Survival
- Heart Transplantation/adverse effects
- Humans
- Immunity, Innate
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Risk Factors
- Signal Transduction
- Treatment Outcome
- Vascular Remodeling
Collapse
Affiliation(s)
- Jordan S Pober
- Department of Immunobiology, Pathology and Dermatology, Yale School of Medicine, 10 Amistad Street, New Haven CT 06520-8089, USA
| | - Sharon Chih
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Jon Kobashigawa
- Department of Medicine, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, USA
| | - Joren C Madsen
- Division of Cardiac Surgery and Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - George Tellides
- Department of Surgery (Cardiac Surgery), Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Habal MV, Miller AM, Rao S, Lin S, Obradovic A, Khosravi-Maharlooei M, See S, Roy P, Ronzon S, Ho SH, Marboe C, Naka Y, Takeda K, Restaino S, Han A, Mancini D, Givertz M, Madsen JC, Sykes M, Addonizio L, Farr M, Zorn E. T cell repertoire analysis suggests a prominent bystander response in human cardiac allograft vasculopathy. Am J Transplant 2021; 21:1465-1476. [PMID: 33021057 PMCID: PMC8672660 DOI: 10.1111/ajt.16333] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 01/25/2023]
Abstract
T cells are implicated in the pathogenesis of cardiac allograft vasculopathy (CAV), yet their clonality, specificity, and function are incompletely defined. Here we used T cell receptor β chain (TCRB) sequencing to study the T cell repertoire in the coronary artery, endomyocardium, and peripheral blood at the time of retransplant in four cases of CAV and compared it to the immunoglobulin heavy chain variable region (IGHV) repertoire from the same samples. High-dimensional flow cytometry coupled with single-cell PCR was also used to define the T cell phenotype. Extensive overlap was observed between intragraft and blood TCRBs in all cases, a finding supported by robust quantitative diversity metrics. In contrast, blood and graft IGHV repertoires from the same samples showed minimal overlap. Coronary infiltrates included CD4+ and CD8+ memory T cells expressing inflammatory (IFNγ, TNFα) and profibrotic (TGFβ) cytokines. These were distinguishable from the peripheral blood based on memory, activation, and tissue residency markers (CD45RO, CTLA-4, and CD69). Importantly, high-frequency rearrangements were traced back to endomyocardial biopsies (2-6 years prior). Comparison with four HLA-mismatched blood donors revealed a repertoire of shared TCRBs, including a subset of recently described cross-reactive sequences. These findings provide supportive evidence for an active local intragraft bystander T cell response in late-stage CAV.
Collapse
Affiliation(s)
- Marlena V. Habal
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY,Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | - April M.I Miller
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Samhita Rao
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Sijie Lin
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Aleksandar Obradovic
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | | | - Sarah See
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Poulomi Roy
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Shihab Ronzon
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Siu-hong Ho
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Charles Marboe
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Yoshifumi Naka
- Department of Surgery, Division of Cardiothoracic Surgery, Columbia University Irving Medical Center, New York, NY
| | - Koji Takeda
- Department of Surgery, Division of Cardiothoracic Surgery, Columbia University Irving Medical Center, New York, NY
| | - Susan Restaino
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | - Arnold Han
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Donna Mancini
- Department of Medicine, Mount Sinai Hospital, Icahn School of Medicine, New York, NY
| | - Michael Givertz
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Joren C. Madsen
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Linda Addonizio
- Department of Pediatrics, Division of Pediatric Cardiology, Columbia University Irving Medical Center, New York, NY
| | - Maryjane Farr
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | - Emmanuel Zorn
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
4
|
Milutinović A, Šuput D, Zorc-Pleskovič R. Pathogenesis of atherosclerosis in the tunica intima, media, and adventitia of coronary arteries: An updated review. Bosn J Basic Med Sci 2020; 20:21-30. [PMID: 31465719 PMCID: PMC7029210 DOI: 10.17305/bjbms.2019.4320] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of arteries and it affects the structure and function of all three layers of the coronary artery wall. Current theories suggest that the dysfunction of endothelial cells is one of the initial steps in the development of atherosclerosis. The view that the tunica intima normally consists of a single layer of endothelial cells attached to the subendothelial layer and internal elastic membrane has been questioned in recent years. The structure of intima changes with age and it becomes multilayered due to migration of smooth muscle cells from the media to intima. At this stage, the migration and proliferation of smooth muscle cells do not cause pathological changes in the intima. The multilayering of intima is classically considered to be an important stage in the development of atherosclerosis, but in fact atherosclerotic plaques develop only focally due to the interplay of various processes that involve the resident and invading inflammatory cells. The tunica media consists of multiple layers of smooth muscle cells that produce the extracellular matrix, and this layer normally does not contain microvessels. During the development of atherosclerosis, the microvessels from the tunica adventitia or from the lumen may penetrate thickened media to provide nutrition and oxygenation. According to some theories, the endothelial dysfunction of these nutritive vessels may significantly contribute to the atherosclerosis of coronary arteries. The adventitia contains fibroblasts, progenitor cells, immune cells, microvessels, and adrenergic nerves. The degree of inflammatory cell infiltration into the adventitia, which can lead to the formation of tertiary lymphoid organs, correlates with the severity of atherosclerotic plaques. Coronary arteries are surrounded by perivascular adipose tissue that also participates in the atherosclerotic process.
Collapse
Affiliation(s)
- Aleksandra Milutinović
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | - Dušan Šuput
- Institute of Pathophysiology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia.
| | - Ruda Zorc-Pleskovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; International Center for Cardiovascular Diseases MC Medicor d.d., Izola, Slovenia.
| |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW The application of regulatory T cell (Treg) therapy in organ transplantation is actively being pursued using unmodified, typically polyclonal cells. As the results of these ongoing clinical trials emerge, it is time to plan the next wave of clinical trials of Tregs. Here we will review a key strategy to improve Treg effectiveness and reduce side effects, namely increasing Treg specificity - both in terms of antigen recognition and localization to the allograft. RECENT FINDINGS Study of chemokine signatures accompanying acute rejection has revealed several chemokines that could be targeted to increase Treg homing. For example, Tregs possessing a Th1-like phenotype and expressing CXCR3 are better able to migrate towards local inflammation. Allografts themselves can be modified to increase Treg-attracting chemokines and Tregs themselves can produce chemokines, facilitating local proximity to their targets of suppression. Finally, tailoring Treg antigen specificity by T-cell or chimeric antigen receptor engineering is another approach to increase the specificity of suppression and optimize localization. SUMMARY Treg localization to the graft is important, but the important role of lymph node and germinal center homing cannot be overlooked. There is an opportunity to learn from advances made in cancer immunotherapy to optimize Treg therapy for transplantation.
Collapse
|
6
|
Zorc-Pleskovič R, Pleskovič A, Vraspir-Porenta O, Zorc M, Milutinović A. Immune cells and vasa vasorum in the tunica media of atherosclerotic coronary arteries. Bosn J Basic Med Sci 2018; 18:240-245. [PMID: 29671719 DOI: 10.17305/bjbms.2018.2951] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/19/2018] [Accepted: 02/19/2018] [Indexed: 11/16/2022] Open
Abstract
In coronary artery disease (CAD), the disruption of the tunica media immune privilege manifests as increased leukocyte infiltration and the formation of vasa vasorum. We aimed to characterize the immune privilege status of the tunica media in human coronary arteries (CAs) with atherosclerotic plaques, by comparing the abundance and composition of immune-cell infiltrates within the individual arterial-wall layers, and by evaluating vasa vasorum neovascularization of the tunica media. The tissue samples were obtained from 36 symptomatic patients with diffuse CAD (aged 60-72 years) who underwent coronary endarterectomy. T and B cells, macrophages and endothelial cells in the CAs were detected by immunohistochemistry. Morphological analysis of CAs showed significant atherosclerotic changes in all specimens. In the media, we observed damage and loss of smooth muscle cells, destruction of the extracellular matrix architecture, and fibrosis. There were 43.3% of immune cells in the intima, 50% in the adventitia, and 6.7% in the media. In the media, 51.1% of the immune cells were T cells (p ˂ 0.001 compared to B cells and macrophages; ANOVA, Scheffe post hoc analysis), 23.5% were B cells, and 25.4% were macrophages. The number of vasa vasorum in the media was 1 in 38.9% of CAs, 2-3 in 36.1%, and ≥4 in 25% of CAs. Our results indicate that, in atherosclerotic CAs, the immune privilege of the media is disrupted by the infiltration of T and B cells, macrophages, and the presence of vasa vasorum.
Collapse
Affiliation(s)
- Ruda Zorc-Pleskovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | | | | | | | | |
Collapse
|
7
|
Huibers MMH, Gareau AJ, Beerthuijzen JMT, Siera-de Koning E, van Kuik J, Kamburova EG, Vink A, de Jonge N, Lee TDG, Otten HG, de Weger RA. Donor-Specific Antibodies Are Produced Locally in Ectopic Lymphoid Structures in Cardiac Allografts. Am J Transplant 2017; 17:246-254. [PMID: 27428759 DOI: 10.1111/ajt.13969] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/17/2016] [Accepted: 07/10/2016] [Indexed: 01/25/2023]
Abstract
Cardiac allograft vasculopathy (CAV) is a transplant pathology, limiting graft survival after heart transplantation. CAV arteries are surrounded by ectopic lymphoid structures (ELS) containing B cells and plasma cells. The aim of this study was to characterize the antigenic targets of antibodies produced in ELS. Coronary arteries and surrounding epicardial tissue from 56 transplant recipients were collected during autopsy. Immunofluorescence was used to identify antibody-producing plasma cells. Immunoglobulin levels in tissue lysates were measured by enzyme-linked immunosorbent assay and analyzed for donor-specific HLA antibodies by Luminex assay. Cytokine and receptor expression levels were quantified using quantitative polymerase chain reaction. Plasma cells in ELS were polyclonal and produced IgG and/or IgM antibodies. In epicardial tissue, IgG (p < 0.05) and IgM levels were higher in transplant patients with larger ELS than smaller ELS. In 4 of 21 (19%) patients with ELS, donor-specific HLA type II antibodies were detected locally. Cytokine and receptor expression (CXCR3, interferon γ and TGF-β) was higher in large ELS in the epicardial tissue than in other vessel wall layers, suggesting active recruitment and proliferation of T and B lymphocytes. ELS exhibited active plasma cells producing locally manufactured antibodies that, in some cases, were directed against the donor HLA, potentially mediating rejection with major consequences for the graft.
Collapse
Affiliation(s)
- M M H Huibers
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - A J Gareau
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - J M T Beerthuijzen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - E Siera-de Koning
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - J van Kuik
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - E G Kamburova
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - A Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - N de Jonge
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - T D G Lee
- Department of Pathology, Dalhousie University, Halifax, Canada.,Department of Surgery, Dalhousie University, Halifax, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
| | - H G Otten
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - R A de Weger
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
8
|
Abstract
Cardiac allograft vasculopathy (CAV) has a high prevalence among patients that have undergone heart transplantation. Cardiac allograft vasculopathy is a multifactorial process in which the immune system is the driving force. In this review, the data on the immunological and fibrotic processes that are involved in the development of CAV are summarized. Areas where a lack of knowledge exists and possible additional research can be completed are pinpointed. During the pathogenesis of CAV, cells from the innate and the adaptive immune system cooperate to reject the foreign heart. This inflammatory response results in dysfunction of the endothelium and migration and proliferation of smooth muscle cells (SMCs). Apoptosis and factors secreted by both the endothelium as well as the SMCs lead to fibrosis. The migration of SMCs together with fibrosis provoke concentric intimal thickening of the coronary arteries, which is the main characteristic of CAV.
Collapse
|
9
|
Abstract
Inflammatory arterial diseases differentially affect the compartments of the vessel wall. The intima and adventitia are commonly involved by the disease process, with luminal and microvascular endothelial cells playing a critical role in the recruitment and activation of leukocytes. In contrast, the avascular media is often spared by immune-mediated disorders. Surprisingly, vascular smooth muscle cells (VSMCs), the predominant and often exclusive cell type of the media, are capable of robust proinflammatory responses to diverse stressors. The multiple cytokines and chemokines produced within the media can profoundly affect macrophage and T cell function, thus amplifying and shaping innate and adaptive immune responses. On the other hand, VSMCs and the extracellular matrix that they produce also display significant anti-inflammatory properties. The balance between the pro- and anti-inflammatory effects of VSMCs and their extracellular matrix versus the strength of the inciting immunologic events determines the pattern of medial pathology. Limitations on the extent of medial infiltration and injury, defined as medial immunoprivilege, are typically seen in arteriosclerotic diseases, such as atherosclerosis and transplant vasculopathy. Conversely, breakdown of medial immunoprivilege that manifests as more intense leukocytic infiltrates, loss of VSMCs, and destruction of the extracellular matrix architecture is a general feature of certain aneurysmal diseases and vasculitides. In this review, we consider the inflammatory and immune functions of VSMCs and how they may lead to medial immunoprivilege or medial inflammation in arterial diseases.
Collapse
Affiliation(s)
- George Tellides
- From the Departments of Surgery (G.T.) and Immunobiology (J.S.P.), Yale University School of Medicine, New Haven, CT; and Veterans Affairs Connecticut Healthcare System, West Haven, CT (G.T.).
| | - Jordan S Pober
- From the Departments of Surgery (G.T.) and Immunobiology (J.S.P.), Yale University School of Medicine, New Haven, CT; and Veterans Affairs Connecticut Healthcare System, West Haven, CT (G.T.)
| |
Collapse
|
10
|
Distinct phenotypes of cardiac allograft vasculopathy after heart transplantation: A histopathological study. Atherosclerosis 2014; 236:353-9. [DOI: 10.1016/j.atherosclerosis.2014.07.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 06/12/2014] [Accepted: 07/17/2014] [Indexed: 11/22/2022]
|
11
|
Zou H, Yang Y, Gao M, Zhang B, Ming B, Sun Y, Chen H, Tang X, Chen Z, Xiong P, Xu Y, Fang M, Tan Z, Gong F, Zheng F. HMGB1 is involved in chronic rejection of cardiac allograft via promoting inflammatory-like mDCs. Am J Transplant 2014; 14:1765-77. [PMID: 24984831 DOI: 10.1111/ajt.12781] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 04/04/2014] [Accepted: 04/06/2014] [Indexed: 01/25/2023]
Abstract
Chronic rejection that leads to diffuse narrowing and occlusion of graft vessels is the most important cause of morbidity and mortality following cardiac transplantation. The role and underlying mechanism of high-mobility group box 1 (HMGB1), as an established inflammatory mediator in acute rejection, remains poorly understood in chronic rejection. Here, we assessed the effects and mechanisms of HMGB1 on the chronic rejection using single MHC Class II-mismatched mouse cardiac transplantation model. It was found that HMGB1 was increased accompanying with the development of chronic rejection, while blockade of HMGB1 with specific neutralizing mAb substantially ameliorated chronic rejection-mediated vasculopathy and fibrosis of allograft, as well as markedly decreased T cell infiltration and production of IL-17A and interferon-gamma in allograft and recipient's spleen. Further, anti-HMGB1 antibody treatment significantly declined the number and frequency of mature dendritic cells (DCs) in allograft and recipient's spleen, especially CD11b(+) Ly6C(high) matured DCs that share the phenotypes with inflammatory-DCs. These findings indicate that HMGB1 contributes to chronic rejection, and HMGB1 blockade may be a novel mean to disrupt the proinflammatory loop after heart transplantation.
Collapse
Affiliation(s)
- H Zou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, China; Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Wang H, Zhang Z, Tian W, Liu T, Han H, Garcia B, Li XC, Du C. Memory T Cells Mediate Cardiac Allograft Vasculopathy and are Inactivated by Anti-OX40L Monoclonal Antibody. Cardiovasc Drugs Ther 2014; 28:115-22. [PMID: 24254032 PMCID: PMC4539019 DOI: 10.1007/s10557-013-6502-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Cardiac allograft vasculopathy (CAV) is a major complication limiting the long-term survival of cardiac transplants. The role of memory T cells (Tmem) in the pathogenesis of CAV remains elusive. This study investigated the role of Tmem cells in the development of CAV and the therapeutic potential of targeting the OX40/OX40L pathway for heart transplant survival. METHODS Tmem cells were generated in Rag-1(-/-) C57BL/6 (B6) mice by homeostatic proliferation (HP) of CD40L null CD3(+) T cells from B6 mice. Rag-1(-/-) B6 mice (H-2(b)) harboring Tmem cells received cardiac allografts from BALB/c mice (H-2(d)), and were either untreated or treated with anti-OX40L monoclonal antibody (mAb) (0.5 mg/mouse/day) for 10 days. RESULTS Six weeks after HP, the majority of transferred CD40L(-/-) T cells in Rag-1(-/-) B6 mice were differentiated to CD44(high) and CD62L(low) Tmem cells. BALB/c heart allografts in Rag-1(-/-) B6 recipient mice in the presence of these Tmem cells developed a typical pathological feature of CAV; intimal thickening, 100 days after transplantation. However, functionally blocking the OX40/OX40L pathway with anti-OX40L mAb significantly prevented CAV development and reduced the Tmem cell population in recipient mice. Anti-OX40L mAb therapy also significantly decreased cellular infiltration and cytokine (IFN-γ, TNF-α and TGF-β) expression in heart allografts. CONCLUSIONS Tmem cells mediate CAV in heart transplants. Functionally blocking the OX40/OX40L pathway using anti-OX40L mAb therapy prevents Tmem cell-mediated CAV, suggesting therapeutic potential for disrupting OX40-OX40L signaling in order to prevent CAV in heart transplant patients.
Collapse
Affiliation(s)
- Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, Tianjin, China,
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Su Y, Jevnikar AM, Huang X, Lian D, Zhang ZX. Spi6 protects alloreactive CD4(+) but not CD8 (+) memory T cell from granzyme B attack by double-negative T regulatory cell. Am J Transplant 2014; 14:580-93. [PMID: 24730048 DOI: 10.1111/ajt.12614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Memory T (Tm) cells pose a major barrier to long-term transplant survival. Whether regulatory T cells (Tregs)can control them remains poorly defined. Previously,we established that double-negative (DN) Tregs suppress effector T (Teff) cells. Here, we demonstrate that DNTregs effectively suppress CD4+/CD8+Teff and CD8+Tm but not CD4+Tm cells, whereas the suppression on CD8+Tm is abrogated by perforin (PFN) deficiency in DNTregs. Consistently, in a BALB/c to B6-Rag1-/-skin transplantation, transfer of DN Tregs suppressed the rejection mediated by CD4þ/CD8+Teff and CD8+Tmcells (76.0±4.9, 87.5±5.0 and 63.0±4.7 days, respectively)but not CD4þTmcells (25.3±1.4 days). Both CD8þ effector memory T and central memory T compartments significantly reduced after DN Treg transfer. CD4+Tm highly expresses granzyme B (GzmB) inhibitor serine protease inhibitor-6 (Spi6). Spi6 deficiency renders CD4þTm susceptible to DN Treg suppression. In addition,transfer of WT DN Tregs, but not PFN-/-DN Tregs,inhibited the skin allograft rejection mediated by Spi6-/-CD4þTm(75.5±7.9 days). In conclusion, CD4+ and CD8+Tm cells differentially respond toDNTregs’ suppression.The GzmB resistance conferred by Spi6 in CD4þTm cells might hint at the physiological significance of Tmpersistence
Collapse
|
14
|
Impact of donor benign intimal thickening on cardiac allograft vasculopathy. J Heart Lung Transplant 2013; 32:454-60. [DOI: 10.1016/j.healun.2013.01.1044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 12/20/2012] [Accepted: 01/22/2013] [Indexed: 11/21/2022] Open
|
15
|
Hogenes MCH, van Dorp S, van Kuik J, Monteiro FRP, ter Hoeve N, van Dijk MR, Martens AC, de Weger RA. Histological assessment of the sclerotic graft-versus-host response in the humanized RAG2-/-γc-/- mouse model. Biol Blood Marrow Transplant 2012; 18:1023-35. [PMID: 22579931 DOI: 10.1016/j.bbmt.2012.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 05/01/2012] [Indexed: 10/28/2022]
Abstract
Graft-versus-host disease (GVHD) remains a frequently occurring and difficult-to-treat complication in human allogeneic stem cell transplantation. Murine transplantation models are often used to study and understand the complex pathogenesis of GVHD and to explore new treatment strategies. Although GVHD kinetics may differ in murine and human models, adequate models are essential for identification of the crucial factors responsible for the major pathology in GVHD. We present a detailed description of the specific histological features of a graft-versus-host-induced fibrotic response in xenogeneic RAG2(-/-)γc(-/-) mice after total body irradiation and injection with human peripheral blood mononuclear cells. We describe the full morphological features of this reaction, including a detailed analysis of the specific tissue infiltration patterns of the human peripheral blood mononuclear cells. Our data show the development of fibrosis, predominantly near blood vessels, and reveal different cell populations and specific cell migration patterns in the affected organs. The combination of immunohistochemical cell characterization and mRNA expression analysis of both human (donor)- and murine (host)-derived cytokines reveals an interaction between host tissues and donor-derived cells in an entangled cytokine profile, in which both donor- and host-derived cytokines contribute to the formation of fibrosis.
Collapse
Affiliation(s)
- Marieke C H Hogenes
- Department of Pathology, University Medical Centre Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Yi T, Fogal B, Hao Z, Tobiasova Z, Wang C, Rao DA, Al-Lamki RS, Kirkiles-Smith NC, Kulkarni S, Bradley JR, Bothwell ALM, Sessa WC, Tellides G, Pober JS. Reperfusion injury intensifies the adaptive human T cell alloresponse in a human-mouse chimeric artery model. Arterioscler Thromb Vasc Biol 2011; 32:353-60. [PMID: 22053072 DOI: 10.1161/atvbaha.111.239285] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Perioperative nonimmune injuries to an allograft can decrease graft survival. We have developed a model for studying this process using human materials. METHODS AND RESULTS Human artery segments were transplanted as infrarenal aortic interposition grafts into an immunodeficient mouse host, allowed to "heal in" for 30 days, and then retransplanted into a second mouse host. To induce a reperfusion injury, the healed-in artery segments were incubated for 3 hours under hypoxic conditions ex vivo before retransplantation. To induce immunologic rejection, the animals receiving the retransplanted artery segment were adoptively transferred with human peripheral blood mononuclear cells or purified T cells from a donor allogeneic to the artery 1 week before surgery. To compare rejection of injured versus healthy tissues, these manipulations were combined. Results were analyzed ex vivo by histology, morphometry, immunohistochemistry, and mRNA quantitation or in vivo by ultrasound. Our results showed that reperfusion injury, which otherwise heals with minimal sequelae, intensifies the degree of allogeneic T cell-mediated injury to human artery segments. CONCLUSIONS We developed a new human-mouse chimeric model demonstrating interactions of reperfusion injury and alloimmunity using human cells and tissues that may be adapted to study other forms of nonimmune injury and other types of adaptive immune responses.
Collapse
Affiliation(s)
- Tai Yi
- Department of Immunobiology, Yale University School of Medicine, 10 Amistad St, New Haven, CT 06520-8089, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Huibers M, De Jonge N, Van Kuik J, Koning ESD, Van Wichen D, Dullens H, Schipper M, De Weger R. Intimal fibrosis in human cardiac allograft vasculopathy. Transpl Immunol 2011; 25:124-32. [PMID: 21782945 DOI: 10.1016/j.trim.2011.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 07/06/2011] [Indexed: 11/30/2022]
Abstract
Human Cardiac Allograft Vasculopathy (CAV) is one of the major complications for patients after heart transplantation. It is characterized by a concentric luminal narrowing due to (neo) intimal expansion in the coronary arteries of donor hearts after heart transplantation. In this process fibrosis plays an important role. Aim of this study is to analyze the factors and cells involved in this fibrotic process. Coronary arteries from five heart transplantation patients and three controls were obtained at autopsy. Quantitative real-time PCR was performed on mRNA obtained from various arterial layers isolated by laser micro dissection. Positive gene expression was confirmed by immunohistochemistry and/or in situ hybridisation. The strongest mRNA expression of fibrotic factors (predominantly pro-fibrotic) was found in the neo-intima. Especially, connective tissue growth factor expression was higher in the CAV vessels than in the controls. The lymphocyte activity of interferon gamma was only detected in CAV vessels. Furthermore as shown by in situ hybridisation, the lymphocytes producing interferon gamma also expressed transforming growth factor beta. Anti-fibrotic factors, such as bone morphogenic protein 4, were only expressed in CD3(-)/CD68(-) stromal cells. Macrophages present in the CAV and control vessels showed to be of the M2 type and did not produce any fibrotic factor(s). In conclusion, T-cells producing both interferon gamma and transforming growth factor beta, may play an important role in the fibrotic process in CAV vessels by upregulation of connective tissue growth factor production.
Collapse
Affiliation(s)
- Manon Huibers
- Department of Pathology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Spatial differences in the presence of FOXP3+ and GranzymeB+ T cells between the intra- and extravascular compartments in renal allograft vasculopathy. PLoS One 2011; 6:e18656. [PMID: 21494640 PMCID: PMC3071842 DOI: 10.1371/journal.pone.0018656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 03/15/2011] [Indexed: 11/25/2022] Open
Abstract
Background Allograft vasculopathy (AV) and native atherosclerosis (NA) share the presence of a T-cell mediated inflammatory response, but differ in overall plaque morphology and growth rate. We studied the distribution and frequency of regulatory- and cytotoxic T cells in the arterial intima lesions in both conditions. Methodology/Principal Findings The study is based on vessels of 15 explanted human renal allografts with AV and 10 carotid artery plaques obtained at surgery. Distribution and frequency of cytotoxic- and regulatory T cells, as identified by the expression of Granzyme B (GrB) and FOXP3 was established in NA and AV. Furthermore, we compared the distribution of these cells in AV with the perivascular, interstitial renal tissue using immunohistochemistry. The total number of T cells was much higher in AV than in NA lesions (711±135 and 37±8 CD3/mm2 respectively, p<0.005, mean, ± SEM). Total numbers of FOXP3+ regulatory cells were also significantly increased in AV (36±10 and 0.9±0.3 FOXP3+/mm2 p<0.05), but relative numbers, expressed as a percentage of the total number of CD3+ T cells ((FOXP3+/CD3+) ×100), were not significantly different (4.6%±0.9 and 2.7%±0.6). GrB+ cells were rare in NA, but significantly increased numbers of GrB+ cells were found in AV lesions (85±24 and 0.2±0.1 GrB+/mm2, p<0.05). Perivascular tissues in the allografts showed a higher relative frequency of FOXP3+ cells than adjacent intimal lesions (14.0%±2.7 and 4.6%±0.9, respectively, p<0.05), but a lower frequency of GrB+ cytotoxic T cells (16.1%±2.7 and 22.6%±3.6, p<0.05). Conclusions Similar to NA, AV is characterized by a low frequency of intimal FOXP3+ regulatory T cells. Moreover, significant spatial differences exist in the distribution of functional T cell subsets between the intra- and extravascular micro-environments of the graft.
Collapse
|
19
|
Wehner JR, Baldwin WM. Cardiac allograft vasculopathy: do adipocytes bridge alloimmune and metabolic risk factors? Curr Opin Organ Transplant 2011; 15:639-44. [PMID: 20689436 DOI: 10.1097/mot.0b013e32833deaee] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW Cardiac allograft vasculopathy (CAV) is still a major cause of chronic graft failure. CAV develops in the coronary arteries as a diffuse, concentric expansion of the intima in conjunction with inflammation and fibrosis of the adventitia. We review recent publications that could link metabolic and immunologic risk factors for CAV.A concept is offered that periarterial adipocytes may provide proinflammatory cytokines that augment immune injury of the coronary arteries. RECENT FINDINGS Clinical and experimental evidence indicate that some alloantibodies and autoantibodies are associated with CAV. Limited data are available on the expression of target antigens on coronary arteries at different times after transplantation. Perivascular adipose tissue is an abundant source of IL-6, IL-8 and MCP-1. Adding to the inflammatory bias, perivascular adipocytes secrete less of the anti-inflammatory adiponectin in comparison to other types of fat. Adiponectin modulates expression of adhesion molecules on the vascular endothelium. It also decreases neointimal formation in arteries following mechanical endovascular injury. SUMMARY Alterations in the balance between proinflammatory and anti-inflammatory cytokines secreted by perivascular fat have been implicated in atherosclerosis and restenosis. This imbalance may also augment the immune responses in the coronary arteries of transplanted hearts.
Collapse
Affiliation(s)
- Jennifer R Wehner
- Department of Immunology NB30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, Ohio, USA
| | | |
Collapse
|
20
|
Zhang M, Wu Q, Shui C. All-trans retinoic acid attenuates cardiac allograft vasculopathy in rats. Transplant Proc 2010; 42:1895-8. [PMID: 20620545 DOI: 10.1016/j.transproceed.2010.02.094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 01/15/2010] [Accepted: 02/26/2010] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We sought to study the inhibitory effects of all-trans retinoic acid (ATRA) on cardiac allograft vasculopathy in rats. METHODS Inbred Wistar and Sprague-Dawley rats were used as donors and recipients, respectively. After abdominal heterotopic heart transplantation, animals were randomized to a cyclosporine (CsA) group versus a CsA+ATRA group: 10 mg/kg/d CsA versus the same CsA dose plus 10 mg/kg/d ATRA. Transplanted hearts were analyzed at 60 days. Cardiac allograft sections were treated with Van Giesson stain to examine vascular luminal occlusion, with immunohistochemistry for CD68 and proliferating cell nuclear antigen (PCNA), and with reverse-transcription polymerase chain reaction (RT-PCR) for platelet-derived growth factor A (PDGF-A) mRNA. RESULTS Luminal occlusion in the CsA+ATRA group was significantly less than that in the CsA group (40.10 +/- 8.20% vs 62.86 +/- 17.18%; P < .01). The CsA+ATRA group showed a marked reduction in PCNA- and CD68-positive cells: namely, 33.96 +/- 8.65% versus 60.17 +/- 17.74% (P < .01) and 17.63 +/- 4.24% versus 32.13 +/- 9.26 (P < .01), respectively. RT-PCR analysis showed that relative PDGF-A mRNA content in the CsA+ATRA group was significantly decreased compared with the CsA group (0.46 +/- 0.08 vs 0.94 +/- 0.11; P < .01). CONCLUSION ATRA may attenuate rat cardiac allograft vasculopathy by inhibiting macrophage infiltration and cell proliferation.
Collapse
Affiliation(s)
- M Zhang
- Department of Cardiovascular Surgery, First Hospital of Tsinghua University, Beijing, China.
| | | | | |
Collapse
|
21
|
Donor HO-1 Expression Inhibits Intimal Hyperplasia in Unmanipulated Graft Recipients: A Potential Role for CD8+ T-Cell Modulation by Carbon Monoxide. Transplantation 2009; 88:653-61. [DOI: 10.1097/tp.0b013e3181b2fd83] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Current world literature. Curr Opin Organ Transplant 2009; 14:103-11. [PMID: 19337155 DOI: 10.1097/mot.0b013e328323ad31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
23
|
Abstract
Transplant vasculopathy (TV) remains the leading cause of late death among heart transplant recipients. Transplant vasculopathy is characterized by progressive neointimal proliferation, leading to ischemic failure of the allograft. Multiple experimental and clinical studies have shown that injury to the graft at various stages of transplantation can be a risk factor for development of transplant vasculopathy. The hallmark of cardiac allograft injury is the infiltration of leukocytes. Recruitment of leukocytes requires intercellular communication between infiltrating cells, endothelium, parenchymal cells, and components of extracellular matrix. These events are mediated via the generation of adhesion molecules, cytokines, and chemokines. The chemokines, by virtue of their specific cell receptor expression, can selectively mediate the local recruitment/activation of distinct leukocytes/cells, allowing for migration across the endothelium and beyond the vascular compartment. This report provides a comprehensive review of the chemokines that participate in the development of transplant vasculopathy.
Collapse
Affiliation(s)
- John A Belperio
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angele, CA 90095, USA
| | | |
Collapse
|