1
|
Suek N, Young T, Fu J. Immune cell profiling in intestinal transplantation. Hum Immunol 2024; 85:110808. [PMID: 38762429 PMCID: PMC11283363 DOI: 10.1016/j.humimm.2024.110808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/20/2024]
Abstract
Since the first published case study of human intestinal transplantation in 1967, there have been significant studies of intestinal transplant immunology in both animal models and humans. An improved understanding of the profiles of different immune cell subsets is critical for understanding their contributions to graft outcomes. While different studies have focused on the contribution of one or a few subsets to intestinal transplant, no study has integrated these data for a comprehensive overview of immune dynamics after intestinal transplant. Here, we provide a systematic review of the literature on different immune subsets and discuss their roles in intestinal transplant outcomes on multiple levels, focusing on chimerism and graft immune reconstitution, clonal alloreactivity, and cell phenotype. In Sections 1, 2 and 3, we lay out a shared framework for understanding intestinal transplant, focusing on the mechanisms of rejection or tolerance in the context of mucosal immunology and illustrate the unique role of the bidirectional graft-versus-host (GvH) and host-versus-graft (HvG) alloresponse. In Sections 4, 5 and 6, we further expand upon these concepts as we discuss the contribution of different cell subsets to intestinal transplant. An improved understanding of intestinal transplantation immunology will bring us closer to maximizing the potential of this important treatment.
Collapse
Affiliation(s)
- Nathan Suek
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Tyla Young
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
2
|
Cohen CD, Rousseau ST, Bermea KC, Bhalodia A, Lovell JP, Dina Zita M, Čiháková D, Adamo L. Myocardial Immune Cells: The Basis of Cardiac Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1198-1207. [PMID: 37068299 PMCID: PMC10111214 DOI: 10.4049/jimmunol.2200924] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/14/2023] [Indexed: 04/19/2023]
Abstract
The mammalian heart is characterized by the presence of striated myocytes, which allow continuous rhythmic contraction from early embryonic development until the last moments of life. However, the myocardium contains a significant contingent of leukocytes from every major class. This leukocyte pool includes both resident and nonresident immune cells. Over recent decades, it has become increasingly apparent that the heart is intimately sensitive to immune signaling and that myocardial leukocytes exhibit an array of critical functions, both in homeostasis and in the context of cardiac adaptation to injury. Here, we systematically review current knowledge of all major leukocyte classes in the heart, discussing their functions in health and disease. We also highlight the connection between the myocardium, immune cells, lymphoid organs, and both local and systemic immune responses.
Collapse
Affiliation(s)
- Charles D. Cohen
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Sylvie T. Rousseau
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Kevin C. Bermea
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Aashik Bhalodia
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jana P. Lovell
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Marcelle Dina Zita
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Luigi Adamo
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
3
|
Fratta Pasini AM, Stranieri C, Busti F, Di Leo EG, Girelli D, Cominacini L. New Insights into the Role of Ferroptosis in Cardiovascular Diseases. Cells 2023; 12:cells12060867. [PMID: 36980208 PMCID: PMC10047059 DOI: 10.3390/cells12060867] [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: 02/13/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the principal cause of disease burden and death worldwide. Ferroptosis is a new form of regulated cell death mainly characterized by altered iron metabolism, increased polyunsaturated fatty acid peroxidation by reactive oxygen species, depletion of glutathione and inactivation of glutathione peroxidase 4. Recently, a series of studies have indicated that ferroptosis is involved in the death of cardiac and vascular cells and has a key impact on the mechanisms leading to CVDs such as ischemic heart disease, ischemia/reperfusion injury, cardiomyopathies, and heart failure. In this article, we reviewed the molecular mechanism of ferroptosis and the current understanding of the pathophysiological role of ferroptosis in ischemic heart disease and in some cardiomyopathies. Moreover, the comprehension of the machinery governing ferroptosis in vascular cells and cardiomyocytes may provide new insights into preventive and therapeutic strategies in CVDs.
Collapse
|
4
|
Li D, Pi W, Sun Z, Liu X, Jiang J. Ferroptosis and its role in cardiomyopathy. Biomed Pharmacother 2022; 153:113279. [PMID: 35738177 DOI: 10.1016/j.biopha.2022.113279] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 12/09/2022] Open
Abstract
Heart disease is the leading cause of death worldwide. Cardiomyopathy is a disease characterized by the heart muscle damage, resulting heart in a structurally and functionally change, as well as heart failure and sudden cardiac death. The key pathogenic factor of cardiomyopathy is the loss of cardiomyocytes, but the related molecular mechanisms remain unclear. Ferroptosis is a newly discovered regulated form of cell death, characterized by iron accumulation and lipid peroxidation during cell death. Recent studies have shown that ferroptosis plays an important regulatory roles in the occurrence and development of many heart diseases such as myocardial ischemia/reperfusion injury, cardiomyopathy and heart failure. However, the systemic association of ferroptosis and cardiomyopathy remains largely unknown and needs to be elucidated. In this review, we provide an overview of the molecular mechanisms of ferroptosis and its role in individual cardiomyopathies, highlight that targeting ferroptosis maybe a potential therapeutic strategy for cardiomyopathy therapy in the future.
Collapse
Affiliation(s)
- Danlei Li
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Zhenzhu Sun
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Xiaoman Liu
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| |
Collapse
|
5
|
Innate Immunity Response to BK Virus Infection in Polyomavirus-Associated Nephropathy in Kidney Transplant Recipients. TRANSPLANTOLOGY 2022. [DOI: 10.3390/transplantology3010003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BK polyomavirus (BKV) mainly causes infection in uroepithelial and renal tubular epithelial cells of either immunocompetent or immunocompromised hosts. Despite asymptomatic or mild clinical features in immunocompetent hosts with BK infection, serious complications are frequently found in immunocompromised patients, especially patients with kidney transplantation. Accordingly, BKV-associated nephropathy (BKVN) demonstrates a wide range of clinical manifestations, including ureteric stenosis and hemorrhagic cystitis. In addition, BKV re-infection in post-kidney transplantation is also a main cause of kidney allograft dysfunction and graft loss. Since the direct anti-BKV is unavailable, immune response against BKV infection is the main mechanism for organism control and might be a novel strategy to treat or suppress BKV. As such, the innate immunity, consisting of immune cells and soluble molecules, does not only suppress BKV but also enhances the subsequent adaptive immunity to eradicate the virus. Furthermore, the re-activation of BKV in BKVN of kidney-transplanted recipients seems to be related to the status of innate immunity. Therefore, this review aims to collate the most recent knowledge of innate immune response against BKV and the association between the innate immunity status of kidney-transplanted recipients and BKV re-activation.
Collapse
|
6
|
Chang Y, Li X, Cheng Q, Hu Y, Chen X, Hua X, Fan X, Tao M, Song J, Hu S. Single-cell transcriptomic identified HIF1A as a target for attenuating acute rejection after heart transplantation. Basic Res Cardiol 2021; 116:64. [PMID: 34870762 DOI: 10.1007/s00395-021-00904-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/04/2021] [Accepted: 11/20/2021] [Indexed: 10/19/2022]
Abstract
Acute rejection (AR) is an important contributor to graft failure, which remains a leading cause of death after heart transplantation (HTX). The regulation of immune metabolism has become a new hotspot in the development of immunosuppressive drugs. In this study, Increased glucose metabolism of cardiac macrophages was found in patients with AR. To find new therapeutic targets of immune metabolism regulation for AR, CD45+ immune cells extracted from murine isografts, allografts, and untransplanted donor hearts were explored by single-cell RNA sequencing. Total 20 immune cell subtypes were identified among 46,040 cells. The function of immune cells in AR were illustrated simultaneously. Cardiac resident macrophages were substantially replaced by monocytes and proinflammatory macrophages during AR. Monocytes/macrophages in AR allograft were more active in antigen presentation and inflammatory recruitment ability, and glycolysis. Based on transcription factor regulation analysis, we found that the increase of glycolysis in monocytes/macrophages was mainly regulated by HIF1A. Inhibition of HIF1A could alleviate inflammatory cells infiltration in AR. To find out the effect of HIF1A on AR, CD45+ immune cells extracted from allografts after HIF1A inhibitor treatment were explored by single-cell RNA sequencing. HIF1A inhibitor could reduce the antigen presenting ability and pro-inflammatory ability of macrophages, and reduce the infiltration of Cd4+ and Cd8a+ T cells in AR. The expression of Hif1α in AR monocytes/macrophages was regulated by pyruvate kinase 2. Higher expression of HIF1A in macrophages was also detected in human hearts with AR. These indicated HIF1A may serve as a potential target for attenuating AR.
Collapse
Affiliation(s)
- Yuan Chang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.,The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xiangjie Li
- School of Statistics and Data Science, Nankai University, Tianjin, 300371, China.,The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Qi Cheng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Ministry of Education, National Health Commission, Wuhan, 430000, China
| | - Yiqing Hu
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xiao Chen
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xiumeng Hua
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xuexin Fan
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Menghao Tao
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Jiangping Song
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China.
| | - Shengshou Hu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.,The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| |
Collapse
|
7
|
Siren EMJ, Luo HD, Tam F, Montgomery A, Enns W, Moon H, Sim L, Rey K, Guan Q, Wang JJ, Wardell CM, Monajemi M, Mojibian M, Levings MK, Zhang ZJ, Du C, Withers SG, Choy JC, Kizhakkedathu JN. Prevention of vascular-allograft rejection by protecting the endothelial glycocalyx with immunosuppressive polymers. Nat Biomed Eng 2021; 5:1202-1216. [PMID: 34373602 DOI: 10.1038/s41551-021-00777-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
Systemic immunosuppression for the mitigation of immune rejection after organ transplantation causes adverse side effects and constrains the long-term benefits of the transplanted graft. Here we show that protecting the endothelial glycocalyx in vascular allografts via the enzymatic ligation of immunosuppressive glycopolymers under cold-storage conditions attenuates the acute and chronic rejection of the grafts after transplantation in the absence of systemic immunosuppression. In syngeneic and allogeneic mice that received kidney transplants, the steric and immunosuppressive properties of the ligated polymers largely protected the transplanted grafts from ischaemic reperfusion injury, and from immune-cell adhesion and thereby immunocytotoxicity. Polymer-mediated shielding of the endothelial glycocalyx following organ procurement should be compatible with clinical procedures for transplant preservation and perfusion, and may reduce the damage and rejection of transplanted organs after surgery.
Collapse
Affiliation(s)
- Erika M J Siren
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Haiming D Luo
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Franklin Tam
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Ashani Montgomery
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Winnie Enns
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Haisle Moon
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Lyann Sim
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin Rey
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Qiunong Guan
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jiao-Jing Wang
- Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA
| | - Christine M Wardell
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Mahdis Monajemi
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Majid Mojibian
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Zheng J Zhang
- Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA
| | - Caigan Du
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen G Withers
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan C Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada. .,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada. .,Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada. .,School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
8
|
Teng Y, Huang Z, Yao L, Wang Y, Li T, Guo J, Wei R, Xia L, Wu Q. Emerging roles of long non-coding RNAs in allotransplant rejection. Transpl Immunol 2021; 70:101408. [PMID: 34015462 DOI: 10.1016/j.trim.2021.101408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/09/2021] [Accepted: 05/14/2021] [Indexed: 01/10/2023]
Abstract
Allotransplantation has extensively been employed for managing end-stage organ failure and malignant tumors. Acute and chronic post-transplant rejections are major causes of late morbidity and mortality after allotransplantation. However, there are no objective diagnostic criteria and specific therapy for post-transplant rejections. Owing to key advances in high-throughput RNA sequencing techniques, a wealth of studies have disclosed that long noncoding RNA (lncRNA) expression increased or decreased evidently in biopsies, blood, plasma, urine and specific cells of rejecting patients, and the dysregulated lncRNAs affected the cellular functions and differentiation of the immune system. Hence, we present an overview of the functions of lncRNAs expressed in various immune cells related to allotransplant rejection. Moreover, our review explores the regulatory interplay of relevant lncRNAs and recipients with or without allograft rejection after solid organ transplantations or hematopoietic stem cell transplantation, then discuss whether these relevant lncRNAs can be molecular biomarkers for diagnosis and new therapeutic targets in the management of post-transplanted patients.
Collapse
Affiliation(s)
- Yao Teng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenli Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lan Yao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yajun Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Guo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruowen Wei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qiuling Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
9
|
Frye CC, Bery AI, Kreisel D, Kulkarni HS. Sterile inflammation in thoracic transplantation. Cell Mol Life Sci 2020; 78:581-601. [PMID: 32803398 DOI: 10.1007/s00018-020-03615-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/20/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
The life-saving benefits of organ transplantation can be thwarted by allograft dysfunction due to both infectious and sterile inflammation post-surgery. Sterile inflammation can occur after necrotic cell death due to the release of endogenous ligands [such as damage-associated molecular patterns (DAMPs) and alarmins], which perpetuate inflammation and ongoing cellular injury via various signaling cascades. Ischemia-reperfusion injury (IRI) is a significant contributor to sterile inflammation after organ transplantation and is associated with detrimental short- and long-term outcomes. While the vicious cycle of sterile inflammation and cellular injury is remarkably consistent amongst different organs and even species, we have begun understanding its mechanistic basis only over the last few decades. This understanding has resulted in the developments of novel, yet non-specific therapies for mitigating IRI-induced graft damage, albeit with moderate results. Thus, further understanding of the mechanisms underlying sterile inflammation after transplantation is critical for identifying personalized therapies to prevent or interrupt this vicious cycle and mitigating allograft dysfunction. In this review, we identify common and distinct pathways of post-transplant sterile inflammation across both heart and lung transplantation that can potentially be targeted.
Collapse
Affiliation(s)
- C Corbin Frye
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA.
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA
| |
Collapse
|
10
|
Sheng L, Mu Q, Wu X, Yang S, Zhu H, Wang J, Lai Y, Wu H, Sun Y, Hu Y, Fu H, Wang Y, Xu K, Sun Y, Zhang Y, Zhang P, Zhou M, Lai B, Xu Z, Gao M, Zhang Y, Ouyang G. Cytotoxicity of Donor Natural Killer Cells to Allo-Reactive T Cells Are Related With Acute Graft-vs.-Host-Disease Following Allogeneic Stem Cell Transplantation. Front Immunol 2020; 11:1534. [PMID: 32849519 PMCID: PMC7411138 DOI: 10.3389/fimmu.2020.01534] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
Objectives: The mechanism and immunoregulatory role of human natural killer (NK) cells in acute graft-vs.-host-disease (aGVHD) remains unclear. This study quantitatively analyzed the cytotoxicity of donor NK cells toward allo-reactive T cells, and investigated their relationship with acute GVHD (aGVHD). Methods: We evaluated NK dose, subgroup, and receptor expression in allografts from 98 patients who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT). A CD107a degranulating assay was used as a quantitative detection method for the cytotoxic function of donor NK cells to allo-reactive T cells. In antibody-blocking assay, NK cells were pre-treated with anti-DNAM-1(CD226), anti-NKG2D, anti-NKP46, or anti-NKG-2A monoclonal antibodies (mAbs) before the degranulating assay. Results: NK cells in allografts effectively inhibited auto-T cell proliferation following alloantigen stimulation, selectively killing alloantigen activated T cells. NKG2A− NK cell subgroups showed higher levels of CD107a degranulation toward activated T cells, when compared with NKG2A− subgroups. Blocking NKG2D or CD226 (DNAM-1) led to significant reductions in degranulation, whereas NKG2A block resulted in increased NK degranulation. Donor NK cells in the aGVHD group expressed lower levels of NKG2D and CD226, higher levels of NKG2A, and showed higher CD107a degranulation levels when compared with NK cells in the non-aGVHD group. Using univariate analysis, higher NK degranulation activities in allografts (CD107ahigh) were correlated with a decreased risk in grade I–IV aGVHD (hazard risk [HR] = 0.294; P < 0.0001), grade III–IV aGVHD (HR = 0.102; P < 0.0001), and relapse (HR = 0.157; P = 0.015), and improved overall survival (HR = 0.355; P = 0.028) after allo-HSCT. Multivariate analyses showed that higher NK degranulation activities (CD107ahigh) in allografts were independent risk factors for grades, I–IV aGVHD (HR = 0.357; P = 0.002), and grades III–IV aGVHD (HR = 0.13; P = 0.009). Conclusions: These findings reveal that the degranulation activity of NK in allografts toward allo-activated T cells was associated with the occurrence and the severity of aGVHD, after allogeneic stem cell transplantation. This suggested that cytotoxicity of donor NK cells to allo-reactive T cells have important roles in aGVHD regulation.
Collapse
Affiliation(s)
- Lixia Sheng
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Qitian Mu
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Xiaoqing Wu
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Shujun Yang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Huiling Zhu
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Jiaping Wang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Yanli Lai
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Hao Wu
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Ye Sun
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huarui Fu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Wang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Kaihong Xu
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Yongcheng Sun
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Yanli Zhang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Ping Zhang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Miao Zhou
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Binbin Lai
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Zhijuan Xu
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Minjie Gao
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Yi Zhang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| |
Collapse
|
11
|
Abstract
The observation that heart failure with reduced ejection fraction is associated with elevated circulating levels of pro-inflammatory cytokines opened a new area of research that has revealed a potentially important role for the immune system in the pathogenesis of heart failure. However, until the publication in 2019 of the CANTOS trial findings on heart failure outcomes, all attempts to target inflammation in the heart failure setting in phase III clinical trials resulted in neutral effects or worsening of clinical outcomes. This lack of positive results in turn prompted questions on whether inflammation is a cause or consequence of heart failure. This Review summarizes the latest developments in our understanding of the role of the innate and adaptive immune systems in the pathogenesis of heart failure, and highlights the results of phase III clinical trials of therapies targeting inflammatory processes in the heart failure setting, such as anti-inflammatory and immunomodulatory strategies. The most recent of these studies, the CANTOS trial, raises the exciting possibility that, in the foreseeable future, we might be able to identify those patients with heart failure who have a cardio-inflammatory phenotype and will thus benefit from therapies targeting inflammation.
Collapse
|
12
|
Ashraf MI, Sarwar A, Kühl AA, Hunger E, Sattler A, Aigner F, Regele H, Sauter M, Klingel K, Schneeberger S, Resch T, Kotsch K. Natural Killer Cells Promote Kidney Graft Rejection Independently of Cyclosporine A Therapy. Front Immunol 2019; 10:2279. [PMID: 31616441 PMCID: PMC6769038 DOI: 10.3389/fimmu.2019.02279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/09/2019] [Indexed: 01/02/2023] Open
Abstract
Natural Killer (NK) cells have recently been recognized as key players in antibody-mediated chronic allograft failure, thus requiring a comprehensive understanding whether NK cells can escape conventional immunosuppressive regimens. Influence of cyclosporine A (CyA) on NK cell function was studied in a mouse model of allogeneic kidney transplantation (KTX, BALB/c to C57BL/6). Recipients were treated daily with CyA (10 mg/kg) for seven or 14 days for long term survival (day 56). Administration of CyA in recipients resulted in significantly reduced frequencies of intragraft and splenic CD8+ T cells, whereas the latter illustrated reduced IFNγ production. In contrast, intragraft and splenic NK cell frequencies remained unaffected in CyA recipients and IFNγ production and degranulation of NK cells were not reduced as compared with controls. Depletion of NK cells in combination with CyA resulted in an improvement in kidney function until day 7 and prolonged graft survival until day 56 as compared to untreated controls. Surviving animals demonstrated higher intragraft frequencies of proliferating CD4+FoxP3+Ki67+ regulatory T (TREG) cells as well as higher frequencies of CD8+CD122+ TREG. We here demonstrate that NK cell depletion combined with CyA synergistically improves graft function and prolongs graft survival, suggesting that NK cell targeting constitutes a novel approach for improving KTX outcomes.
Collapse
Affiliation(s)
| | - Attia Sarwar
- Department of General, Visceral and Vascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja A Kühl
- iPath.Berlin-Immunopathology for Experimental Models, Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Elena Hunger
- Department of General, Visceral and Vascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Arne Sattler
- Department of General, Visceral and Vascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Aigner
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heinz Regele
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Martina Sauter
- Department of Molecular Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Karin Klingel
- Department of Molecular Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Resch
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Katja Kotsch
- Department of General, Visceral and Vascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
13
|
Beetz O, Kolb J, Buck B, Trautewig B, Timrott K, Vondran FWR, Meder I, Löbbert C, Hundrieser J, Klempnauer J, Bektaş H, Lieke T. Recipient natural killer cells alter the course of rejection of allogeneic heart grafts in rats. PLoS One 2019; 14:e0220546. [PMID: 31437165 PMCID: PMC6705777 DOI: 10.1371/journal.pone.0220546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
Rejection of solid organ grafts is regarded to be dependent on T cell responses. Nonetheless, numerous studies have focused on the contribution of NK cells in this process, resulting in contradictory theories. While some conclude that there is no participation of NK cells, others found an inflammatory or regulative role of NK cells. However, the experimental settings are rarely comparable with regard to challenged species, strain combinations or the nature of the graft. Thus, clear definition of NK cell contribution might be impeded by these circumstances. In this study we performed heterotopic heart transplantation (HTx) in rats, choosing one donor-recipient-combination leading to a fast and a second leading to a prolonged course of graft rejection. We intervened in the rejection process, by depletion of recipient NK cells on the one hand and by injection of activated NK cells syngeneic to the recipients on the other. The fast course of rejection could not be influenced by any of the NK cell manipulative treatments. However, the more prolonged course of rejection was highly susceptible to depletion of NK cells, resulting in significant acceleration of rejection, while injection of NK cells induced acceptance of the grafts. We suggest that, depending on the specific setting, NK cells can attenuate the first trigger of immune response, which allows establishing the regulatory activity leading to tolerance of the graft.
Collapse
Affiliation(s)
- Oliver Beetz
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Joline Kolb
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Benjamin Buck
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Britta Trautewig
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
- Transplant Laboratory, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Kai Timrott
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Florian W. R. Vondran
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Ingrid Meder
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Corinna Löbbert
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Joachim Hundrieser
- Transplant Laboratory, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Jürgen Klempnauer
- Transplant Laboratory, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Hüseyin Bektaş
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
- Department of General-, Visceral- and Oncological Surgery, Hospital Group Gesundheit Nord, Bremen, Germany
| | - Thorsten Lieke
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
14
|
Adithan A, John Peter JS, Hossain MA, Kang CW, Kim B, Kim NS, Hwang KC, Kim JH. Biological effects of cyclosporin A on CD3 -CD161 + and CD3 +CD161 + lymphocytes. Mol Cell Biochem 2019; 458:159-169. [PMID: 31020492 DOI: 10.1007/s11010-019-03539-2] [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: 11/23/2018] [Accepted: 04/12/2019] [Indexed: 11/24/2022]
Abstract
Cyclosporin A (CSA) is a widely used drug to prevent the immune cell function. It is well known that CSA blocks transcription of cytokine genes in activated T cells. The connection between T cells and CSA has been well established. However, the effect of CSA on natural killer (NK) cells is not thoroughly understood. Therefore, in the present study, splenocytes and peripheral blood mononuclear cells (PBMCs) were treated with CSA in the presence of concanavalin A (Con A) or interleukin-2 (IL-2). CSA at higher concentrations induces apoptosis and inhibition of proliferation, while lower concentrations showed synergistically enhanced proliferation in splenocytes and PBMCs. Further, CSA favored the in vitro conversion of CD3+CD161+ cells. Splenocytes and PBMC were found to have synergistic proliferation with Con A, and PBMC exhibited significantly higher expression of NKp30, NKp44, and granzyme B along with enhanced cytotoxicity against K-562 cells in CSA-treated animals. Proliferation assay also showed that proliferation of CD161+ cells was higher in CSA-treated animals. Collectively, our results suggest that CSA differentially influences the population, function, and expression of the NK cell phenotype.
Collapse
Affiliation(s)
- Aravinthan Adithan
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan, Jeollabuk-Do, Republic of Korea
| | - Judith Sharmila John Peter
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan, Jeollabuk-Do, Republic of Korea
| | - Mohammad Amjad Hossain
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan, Jeollabuk-Do, Republic of Korea
| | - Chang-Won Kang
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan, Jeollabuk-Do, Republic of Korea
| | - Bumseok Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan, Jeollabuk-Do, Republic of Korea
| | - Nam Soo Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan, Jeollabuk-Do, Republic of Korea
| | - Ki-Chul Hwang
- Department of Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan, Jeollabuk-Do, Republic of Korea.
| |
Collapse
|
15
|
Tissue-Resident Lymphocytes in Solid Organ Transplantation: Innocent Passengers or the Key to Organ Transplant Survival? Transplantation 2018; 102:378-386. [PMID: 29135830 DOI: 10.1097/tp.0000000000002001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Short-term outcomes of solid organ transplantation have improved dramatically over the past several decades; however, long-term survival has remained static over the same period, and chronic rejection remains a major cause of graft failure. The importance of donor, or "passenger," lymphocytes to the induction of tolerance to allografts was recognized in the 1990s, but their precise contribution to graft acceptance or rejection has not been elucidated. Recently, specialized populations of tissue-resident lymphocytes in nonlymphoid organs have been described. These lymphocytes include tissue-resident memory T cells, regulatory T cells, γδ T cells, invariant natural killer T cells, and innate lymphoid cells. These cells reside in commonly transplanted solid organs, including the liver, kidneys, heart, and lung; however, their contribution to graft acceptance or rejection has not been examined in detail. Similarly, it is unclear whether tissue-resident cells derived from the pool of recipient-derived lymphocytes play a specific role in transplantation biology. This review summarizes the evidence for the roles of tissue-resident lymphocytes in transplant immunology, focussing on their features, functions, and relevance for solid organ transplantation, with specific reference to liver, kidney, heart, and lung transplantation.
Collapse
|
16
|
Javaheri A, Wang AR, Luning Prak E, Lal P, Goldberg LR, Kamoun M. Fatal accelerated rejection with a prominent natural killer cell infiltrate in a heart transplant recipient with peripartum cardiomyopathy. Transpl Immunol 2017; 47:49-54. [PMID: 29101003 DOI: 10.1016/j.trim.2017.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 10/18/2022]
Abstract
Accelerated rejection is uncommon after cardiac transplantation. The mechanism is hypothesized to be mediated by cytotoxic T cells and anti-HLA antibodies resulting from a memory response to the donor allograft in sensitized patients. A role for Natural Killer (NK) cell in cellular rejection has also been suggested. We report a case of fulminant accelerated rejection in a heart transplant recipient, with a history of peripartum cardiomyopathy (PPMC). The patient had no pre-transplant donor specific antibody and flow cytometric T and B cell crossmatches were negative. Autopsy revealed left ventricular subendocardial and intramyocardial hemorrhage with diffuse lymphocytic infiltrates and myocyte damage (Grade 3R rejection). Immunohistochemistry revealed a large proportion (50-70%) of mature CD16+ NK cells with cytotoxic potential in the interstitium and the intra capillary compartments. This case highlights the need for evaluating the potential role of NK cells in accelerated rejection in heart transplant recipients.
Collapse
Affiliation(s)
- Ali Javaheri
- Washington University School of Medicine, Division of Cardiology, United States
| | - Amber R Wang
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, United States
| | - Eline Luning Prak
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, United States
| | - Priti Lal
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, United States
| | - Lee R Goldberg
- University of Pennsylvania, Division of Cardiology, Department of Medicine, United States
| | - Malek Kamoun
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, United States.
| |
Collapse
|
17
|
Shimata K, Sakamoto R, Anan T, Uchida K, Honda M, Kouroki M, Urabe T, Hayashida S, Yamamoto H, Sugawara Y, Inomata Y. Fatal graft-versus-host disease after living-donor liver transplantation from an HLA-DR-mismatched donor. Pediatr Transplant 2017; 21. [PMID: 28834141 DOI: 10.1111/petr.13039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/27/2017] [Indexed: 11/30/2022]
Abstract
Acute GVHD is a rare complication after liver transplantation that has a high mortality rate. We experienced an infant case complicated with acute GVHD. An 8-month-old infant with biliary atresia underwent LDLT with a graft obtained from his mother. Their HLAs showed a donor-dominant one-way match, not at HLA-DR but at HLA-A, HLA-B, and HLA-C (recipient; A 31/33, B 51/54, C 1/14, DR 9/11, donor; A 31/-, B 51/-, C 14/-, DR 8/11). The patient exhibited a high fever, skin rash, and diarrhea, and was diagnosed with acute GVHD based on the blood chimerism test. Despite immunosuppression treatment with prednisolone and tacrolimus, plasma exchange, blood transfusion including cord blood transplantation, and antibiotics, the child died on postoperative day 126. Donor-dominant one-way matching at HLA class 1 can be a high-risk factor for acute GVHD despite HLA class 2 mismatching.
Collapse
Affiliation(s)
- Keita Shimata
- Department of Transplantation and Pediatric Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Rieko Sakamoto
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
| | - Tadashi Anan
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
| | - Koushi Uchida
- Department of Transplantation and Pediatric Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Masaki Honda
- Department of Transplantation and Pediatric Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Masahiko Kouroki
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
| | - Tomonari Urabe
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
| | - Shintaro Hayashida
- Department of Transplantation and Pediatric Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Hidekazu Yamamoto
- Department of Transplantation and Pediatric Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Yasuhiko Sugawara
- Department of Transplantation and Pediatric Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Yukihiro Inomata
- Department of Transplantation and Pediatric Surgery, Kumamoto University Hospital, Kumamoto, Japan
| |
Collapse
|
18
|
Mahr B, Wekerle T. Murine models of transplantation tolerance through mixed chimerism: advances and roadblocks. Clin Exp Immunol 2017; 189:181-189. [PMID: 28395110 PMCID: PMC5508343 DOI: 10.1111/cei.12976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2017] [Indexed: 02/06/2023] Open
Abstract
Organ transplantation is the treatment of choice for patients with end-stage organ failure, but chronic immunosuppression is taking its toll in terms of morbidity and poor efficacy in preventing late graft loss. Therefore, a drug-free state would be desirable where the recipient permanently accepts a donor organ while remaining otherwise fully immunologically competent. Mouse studies unveiled mixed chimerism as an effective approach to induce such donor-specific tolerance deliberately and laid the foundation for a series of clinical pilot trials. Nevertheless, its widespread clinical implementation is currently prevented by cytotoxic conditioning and limited efficacy. Therefore, the use of mouse studies remains an indispensable tool for the development of novel concepts with potential for translation and for the delineation of underlying tolerance mechanisms. Recent innovations developed in mice include the use of pro-apoptotic drugs or regulatory T cell (Treg ) transfer for promoting bone marrow engraftment in the absence of myelosuppression and new insight gained in the role of innate immunity and the interplay between deletion and regulation in maintaining tolerance in chimeras. Here, we review these and other recent advances in murine studies inducing transplantation tolerance through mixed chimerism and discuss both the advances and roadblocks of this approach.
Collapse
Affiliation(s)
- B. Mahr
- Section of Transplantation Immunology, Department of SurgeryMedical University of ViennaViennaAustria
| | - T. Wekerle
- Section of Transplantation Immunology, Department of SurgeryMedical University of ViennaViennaAustria
| |
Collapse
|
19
|
Tsoneva D, Minev B, Frentzen A, Zhang Q, Wege AK, Szalay AA. Humanized Mice with Subcutaneous Human Solid Tumors for Immune Response Analysis of Vaccinia Virus-Mediated Oncolysis. MOLECULAR THERAPY-ONCOLYTICS 2017; 5:41-61. [PMID: 28480327 PMCID: PMC5415323 DOI: 10.1016/j.omto.2017.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/02/2017] [Indexed: 12/20/2022]
Abstract
Oncolytic vaccinia virus (VACV) therapy is an alternative cancer treatment modality that mediates targeted tumor destruction through a tumor-selective replication and an induction of anti-tumor immunity. We developed a humanized tumor mouse model with subcutaneous human tumors to analyze the interactions of VACV with the developing tumors and human immune system. A successful systemic reconstitution with human immune cells including functional T cells as well as development of tumors infiltrated with human T and natural killer (NK) cells was observed. We also demonstrated successful in vivo colonization of such tumors with systemically administered VACVs. Further, a new recombinant GLV-1h376 VACV encoding for a secreted human CTLA4-blocking single-chain antibody (CTLA4 scAb) was tested. Surprisingly, although proving CTLA4 scAb's in vitro binding ability and functionality in cell culture, beside the significant increase of CD56bright NK cell subset, GLV-1h376 was not able to increase cytotoxic T or overall NK cell levels at the tumor site. Importantly, the virus-encoded β-glucuronidase as a measure of viral titer and CTLA4 scAb amount was demonstrated. Therefore, studies in our "patient-like" humanized tumor mouse model allow the exploration of newly designed therapy strategies considering the complex relationships between the developing tumor, the oncolytic virus, and the human immune system.
Collapse
Affiliation(s)
- Desislava Tsoneva
- Department of Biochemistry, Biocenter, University of Wuerzburg, 97074 Wuerzburg, Germany
| | - Boris Minev
- Department of Radiation Medicine and Applied Sciences, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA.,Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA
| | - Alexa Frentzen
- Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA
| | - Qian Zhang
- Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA
| | - Anja K Wege
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Aladar A Szalay
- Department of Biochemistry, Biocenter, University of Wuerzburg, 97074 Wuerzburg, Germany.,Department of Radiation Medicine and Applied Sciences, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA.,Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA.,Rudolph Virchow Center for Experimental Biomedicine, University of Wuerzburg, 97080 Wuerzburg, Germany
| |
Collapse
|
20
|
Risti M, Bicalho MDG. MICA and NKG2D: Is There an Impact on Kidney Transplant Outcome? Front Immunol 2017; 8:179. [PMID: 28289413 PMCID: PMC5326783 DOI: 10.3389/fimmu.2017.00179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/07/2017] [Indexed: 01/06/2023] Open
Abstract
This paper aims to present an overview of MICA and natural killer group 2 member D (NKG2D) genetic and functional interactions and their impact on kidney transplant outcome. Organ transplantation has gone from what can accurately be called a “clinical experiment” to a routine and reliable practice, which has proven to be clinically relevant, life-saving and cost-effective when compared with non-transplantation management strategies of both chronic and acute end-stage organ failures. The kidney is the most frequently transplanted organ in the world (transplant-observatory1). The two treatment options for end-stage renal disease (ESRD) are dialysis and/or transplantation. Compared with dialysis, transplantation is associated with significant improvements in quality of life and overall longevity. A strong relationship exists between allograft loss and human leukocyte antigens (HLA) antibodies (Abs). HLA Abs are not the only factor involved in graft loss, as multiple studies have shown that non-HLA antigens are also involved, even when a patient has a good HLA matche and receives standard immunosuppressive therapy. A deeper understanding of other biomarkers is therefore important, as it is likely to lead to better monitoring (and consequent success) of organ transplants. The objective is to fill the void left by extensive reviews that do not often dive this deep into the importance of MICA and NKG2D in allograft acceptance and their partnership in the immune response. There are few papers that explore the relationship between these two protagonists when it comes to kidney transplantation. This is especially true for the role of NKG2D in kidney transplantation. These reasons give a special importance to this review, which aims to be a helpful tool in the hands of researchers in this field.
Collapse
Affiliation(s)
- Matilde Risti
- LIGH - Immunogenetics and Histocompatibility Laboratory, Federal University of Paraná , Curitiba , Brazil
| | - Maria da Graça Bicalho
- LIGH - Immunogenetics and Histocompatibility Laboratory, Federal University of Paraná , Curitiba , Brazil
| |
Collapse
|
21
|
Rajalingam R. The Impact of HLA Class I-Specific Killer Cell Immunoglobulin-Like Receptors on Antibody-Dependent Natural Killer Cell-Mediated Cytotoxicity and Organ Allograft Rejection. Front Immunol 2016; 7:585. [PMID: 28066408 PMCID: PMC5165035 DOI: 10.3389/fimmu.2016.00585] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/25/2016] [Indexed: 12/21/2022] Open
Abstract
Natural killer (NK) cells of the innate immune system are cytotoxic lymphocytes that play an important roles following transplantation of solid organs and hematopoietic stem cells. Recognition of self-human leukocyte antigen (HLA) class I molecules by inhibitory killer cell immunoglobulin-like receptors (KIRs) is involved in the calibration of NK cell effector capacities during the developmental stage, allowing the subsequent recognition and elimination of target cells with decreased expression of self-HLA class I (due to virus infection or tumor transformation) or HLA class I disparities (in the setting of allogeneic transplantation). NK cells expressing an inhibitory KIR-binding self-HLA can be activated when confronted with allografts lacking a ligand for the inhibitory receptor. Following the response of the adaptive immune system, NK cells can further destroy allograft endothelium by antibody-dependent cell-mediated cytotoxicity (ADCC), triggered through cross-linking of the CD16 Fc receptor by donor-specific antibodies bound to allograft. Upon recognizing allogeneic target cells, NK cells also secrete cytokines and chemokines that drive maturation of dendritic cells to promote cellular and humoral adaptive immune responses against the allograft. The cumulative activating and inhibitory signals generated by ligation of the receptors regulates mature NK cell killing of target cells and their production of cytokines and chemokines. This review summarizes the role of NK cells in allograft rejection and proposes mechanistic concepts that indicate a prominent role for KIR-HLA interactions in facilitating NK cells for Fc receptor-mediated ADCC effector function involved in antibody-mediated rejection of solid organ transplants.
Collapse
Affiliation(s)
- Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco , San Francisco, CA , USA
| |
Collapse
|
22
|
Natural killer cells in inflammatory heart disease. Clin Immunol 2016; 175:26-33. [PMID: 27894980 DOI: 10.1016/j.clim.2016.11.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/09/2016] [Accepted: 11/20/2016] [Indexed: 02/07/2023]
Abstract
Despite of a multitude of excellent studies, the regulatory role of natural killer (NK) cells in the pathogenesis of inflammatory cardiac disease is greatly underappreciated. Clinical abnormalities in the numbers and functions of NK cells are observed in myocarditis and inflammatory dilated cardiomyopathy (DCMi) as well as in cardiac transplant rejection [1-6]. Because treatment of these disorders remains largely symptomatic in nature, patients have little options for targeted therapies [7,8]. However, blockade of NK cells and their receptors can protect against inflammation and damage in animal models of cardiac injury and inflammation. In these models, NK cells suppress the maturation and trafficking of inflammatory cells, alter the local cytokine and chemokine environments, and induce apoptosis in nearby resident and hematopoietic cells [1,9,10]. This review will dissect each protective mechanism employed by NK cells and explore how their properties might be exploited for their therapeutic potential.
Collapse
|
23
|
Shibasaki Y, Hatanaka C, Matsuura Y, Miyazawa R, Yabu T, Moritomo T, Nakanishi T. Effects of IFNγ administration on allograft rejection in ginbuna crucian carp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 62:108-115. [PMID: 27156851 DOI: 10.1016/j.dci.2016.04.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/29/2016] [Accepted: 04/29/2016] [Indexed: 06/05/2023]
Abstract
In vertebrates, the rejection of allografts is primarily accomplished by cell-mediated immunity. We recently identified four IFNγ isoforms with antiviral activity in ginbuna crucian carp, Carassius auratus langsdorfii. However, involvement of the IFNγ isoforms in cell-mediated immunity, especially in T cell function remains unknown. Here we investigate expression of the IFNγ isoforms and effects of administration of recombinant IFNγ (rgIFNγ) isoforms in ginbuna scale allograft rejection. All four IFNγ isoforms showed significantly higher expression with the progression of graft rejection. Administration of rgIFNγrel 1 but not rgIFNγrel 2, rgIFNγ1 nor rgIFNγ2 enhanced allograft rejection. The number of CD4(+) and CD8α(+) cells increased in early stages of rejection, while sIgM(+) cells were higher than controls at day 0 and 5 in the rgIFNγrel 1 administrated group. Expression of IFNγ1 and IFNγ2 mRNA was significantly up-regulated by rgIFNγrel 1 administration, while that of IFNγrel 1 and IFNγrel 2 was not. These results suggest different contributions of the four IFNγ isoforms toward the immune responses comprising allograft rejection.
Collapse
Affiliation(s)
- Yasuhiro Shibasaki
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Chihiro Hatanaka
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Yuta Matsuura
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Ryuichiro Miyazawa
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Takeshi Yabu
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Tadaaki Moritomo
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Teruyuki Nakanishi
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-0880, Japan.
| |
Collapse
|
24
|
Frenkel D, Zhang F, Guirnalda P, Haynes C, Bockstal V, Radwanska M, Magez S, Black SJ. Trypanosoma brucei Co-opts NK Cells to Kill Splenic B2 B Cells. PLoS Pathog 2016; 12:e1005733. [PMID: 27403737 PMCID: PMC4942092 DOI: 10.1371/journal.ppat.1005733] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/08/2016] [Indexed: 11/19/2022] Open
Abstract
After infection with T. brucei AnTat 1.1, C57BL/6 mice lost splenic B2 B cells and lymphoid follicles, developed poor parasite-specific antibody responses, lost weight, became anemic and died with fulminating parasitemia within 35 days. In contrast, infected C57BL/6 mice lacking the cytotoxic granule pore-forming protein perforin (Prf1-/-) retained splenic B2 B cells and lymphoid follicles, developed high-titer antibody responses against many trypanosome polypeptides, rapidly suppressed parasitemia and did not develop anemia or lose weight for at least 60 days. Several lines of evidence show that T. brucei infection-induced splenic B cell depletion results from natural killer (NK) cell-mediated cytotoxicity: i) B2 B cells were depleted from the spleens of infected intact, T cell deficient (TCR-/-) and FcγRIIIa deficient (CD16-/-) C57BL/6 mice excluding a requirement for T cells, NKT cell, or antibody-dependent cell-mediated cytotoxicity; ii) administration of NK1.1 specific IgG2a (mAb PK136) but not irrelevant IgG2a (myeloma M9144) prevented infection-induced B cell depletion consistent with a requirement for NK cells; iii) splenic NK cells but not T cells or NKT cells degranulated in infected C57BL/6 mice co-incident with B cell depletion evidenced by increased surface expression of CD107a; iv) purified NK cells from naïve C57BL/6 mice killed purified splenic B cells from T. brucei infected but not uninfected mice in vitro indicating acquisition of an NK cell activating phenotype by the post-infection B cells; v) adoptively transferred C57BL/6 NK cells prevented infection-induced B cell population growth in infected Prf1-/- mice consistent with in vivo B cell killing; vi) degranulated NK cells in infected mice had altered gene and differentiation antigen expression and lost cytotoxic activity consistent with functional exhaustion, but increased in number as infection progressed indicating continued generation. We conclude that NK cells in T. brucei infected mice kill B cells, suppress humoral immunity and expedite early mortality.
Collapse
Affiliation(s)
- Deborah Frenkel
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Fengqiu Zhang
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Patrick Guirnalda
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Carole Haynes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Viki Bockstal
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Stefan Magez
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Structural Biology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Samuel J. Black
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
25
|
Seyda M, Elkhal A, Quante M, Falk CS, Tullius SG. T Cells Going Innate. Trends Immunol 2016; 37:546-556. [PMID: 27402226 DOI: 10.1016/j.it.2016.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023]
Abstract
Natural killer (NK) cell receptors (NKRs) play a crucial role in the homeostasis of antigen-experienced T cells. Indeed, prolonged antigen stimulation may induce changes in the receptor repertoire of T cells to a profile that features NKRs. Chronic antigen exposure, at the same time, has been shown to trigger the loss of costimulatory CD28 molecules with recently reported intensified antigen thresholds of antigen-experienced CD8(+) T cells. In transplantation, NKRs have been shown to assist allograft rejection in a CD28-independent fashion. We discuss here a role for CD28-negative T cells that have acquired the competency of the NKR machinery, potentially promoting allorecognition either through T cell receptor (TCR) crossreactivity or independently from TCR recognition. Collectively, NKRs can bring about innate-like T cells by providing alternative costimulatory pathways that gain relevance in chronic inflammation, potentially leading to resistance to CD28-targeting immunosuppressants.
Collapse
Affiliation(s)
- Midas Seyda
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - Abdallah Elkhal
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Markus Quante
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christine S Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Hannover, Germany
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Yu X, Lu L, Liu Z, Yang T, Gong X, Ning Y, Jiang Y. Brain-derived neurotrophic factor modulates immune reaction in mice with peripheral nerve xenotransplantation. Neuropsychiatr Dis Treat 2016; 12:685-94. [PMID: 27099498 PMCID: PMC4820192 DOI: 10.2147/ndt.s98387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) has been demonstrated to play an important role in survival, differentiation, and neurite outgrowth for many types of neurons. This study was designed to identify the role of BDNF during peripheral nerve xenotransplantation. MATERIALS AND METHODS A peripheral nerve xenotransplantation from rats to mice was performed. Intracellular cytokines were stained for natural killer (NK) cells, natural killer T (NKT) cells, T cells, and B cells and analyzed by flow cytometry in the spleen of the recipient mouse. Serum levels of related cytokines were quantified by cytometric bead array. RESULTS Splenic NK cells significantly increased in the xenotransplanted mice (8.47±0.88×10(7) cells/mL) compared to that in the control mice (4.66±0.78×10(7) cells/mL, P=0.0003), which significantly reduced in the presence of BDNF (4.85±0.87×10(7) cells/mL, P=0.0004). In contrast, splenic NKT cell number was significantly increased in the mice with xenotransplantation plus BDNF (XT + BDNF) compared to that of control group or of mice receiving xenotransplantation only (XT only). Furthermore, the number of CD3+ T cells, CD3+CD4+ T cells, CD3+CD4- T cells, interferon-γ-producing CD3+CD4+ T cells, and interleukin (IL)-17-producing CD3+CD4+ T cells, as well as CD3-CD19+ B cells, was significantly higher in the spleen of XT only mice compared to the control mice (P<0.05), which was significantly reduced by BDNF (P<0.05). The number of IL-4-producing CD3+CD4+ T cells and CD3+CD4+CD25+Foxp3+ T cells was significantly higher in the spleen of XT + BDNF mice than that in the spleen of XT only mice (P<0.05). Serum levels of IL-6, TNF-α, interferon-γ, and IL-17 were decreased, while IL-4 and IL-10 were stimulated by BDNF following xenotransplantation. CONCLUSION BDNF reduced NK cells but increased NKT cell accumulation in the spleen of xenotransplanted mice. BDNF modulated the number of splenic T cells and its subtype cells in the mice following xenotransplantation. These findings suggest that BDNF inhibits rejection of peripheral nerve following xenotransplantation by regulating innate as well as adaptive immune reaction.
Collapse
Affiliation(s)
- Xin Yu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Laijin Lu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhigang Liu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Teng Yang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xu Gong
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Yubo Ning
- Department of Orthopedics, Ningshi Orthopedics Hospital of Tonghua, Tonghua, People's Republic of China
| | - Yanfang Jiang
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
27
|
Eggenhofer E, Sabet-Rashedi M, Lantow M, Renner P, Rovira J, Koehl GE, Schlitt HJ, Geissler EK, Kroemer A. RORγt(+) IL-22-producing NKp46(+) cells protect from hepatic ischemia reperfusion injury in mice. J Hepatol 2016; 64:128-34. [PMID: 26341825 DOI: 10.1016/j.jhep.2015.08.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/30/2015] [Accepted: 08/14/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS NKp46(+) cells are major effector cells in the pathogenesis of hepatic ischemia reperfusion injury (IRI). Nevertheless, the precise role of unconventional subsets like the IL-22-producing NKp46(+) cells (NK22) remains unknown. The purpose of this study was to examine the role of NK22 cells in IRI in transplantation, particularly with respect to regulation by the transcription factor ROR-gamma-t (RORγt). METHODS To explore the role of NK22 cells in IRI in the absence of adaptive immunity, B6.RORγt-(gfp/wt)-reporter and B6.RORγt-(gfp/gfp)-knockout (KO) mice on a Rag KO background underwent 90min partial warm ischemia, followed by 24h of reperfusion. RESULTS Rag KO mice that possess fully functional NKp46(+) cells, and Rag-common-γ-chain-double-KO (Rag-γc-DKO) mice that lack T, B and NKp46(+) cells, were used as controls. We found that Rag-γc-DKO mice lacking NK22 cells show more severe levels of hepatocellular damage (GPT, histological injury) when compared to both Rag-RORγt-reporter and Rag KO mice that possess NK22 cells. Importantly, Rag-RORγt-reporter and Rag KO mice undergoing IRI expressed high protein levels of both IL-22 and GFP (RORγt), suggesting a protective role for RORγt(+) NK22 cells in IRI. Therefore, we tested the hypothesis that RORγt critically protects from IRI through the induction of hepatic NK22 cells by studying Rag-Rorγt-DKO mice under IRI conditions. We found that the lack of RORγt(+) NK22 cells in Rag-Rorγt-DKO mice significantly enhanced IR-induced hepatocellular injury, a phenotype that could be reversed upon adoptive transfer of Rag-Rorγt-reporter NK22 cells into DKO mice. CONCLUSIONS RORγt(+) NK22 cells play an important protective role in IRI in mice.
Collapse
Affiliation(s)
- Elke Eggenhofer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany.
| | | | - Margareta Lantow
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Philipp Renner
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jordi Rovira
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Gudrun E Koehl
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Alexander Kroemer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany; MedStar Georgetown Transplant Institute, Georgetown University Hospital, Washington, DC, USA.
| |
Collapse
|
28
|
Role of innate immunity in primary graft dysfunction after lung transplantation. Curr Opin Organ Transplant 2015; 18:518-23. [PMID: 23995372 DOI: 10.1097/mot.0b013e3283651994] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Primary graft dysfunction (PGD), a form of acute lung injury after lung transplantation, has a significant impact on clinical outcomes after lung transplantation. This potentially reversible graft impairment occurs after ischemia-reperfusion injury. This review describes the expanding body of literature evaluating the central role of innate immune activation, nonadaptive responses and dysregulation in the development of PGD after lung transplant. RECENT FINDINGS The innate immune system, highlighted by Toll-like receptor pathways and neutrophil migration and influx, plays an important role in the initiation and propagation of ischemia-reperfusion injury. Recent plasma biomarker and gene association studies have identified several genes and proteins composing innate immune pathways to be associated with PGDs. Long pentraxin-3 and Toll-like receptors, as well as inflammasomes and Toll-interacting protein, are associated with the development of PGD after lung transplantation. SUMMARY Innate immune pathways are involved in the development of PGD and may provide attractive targets for therapies. It may be possible to prevent or treat PGD, as well as to allow pre-transplant PGD risk stratification. To improve understanding of the mechanisms behind clinical risk factors for PGD will require further in-depth correlation of donor-specific and recipient-related triggers of nonadaptive immune responses.
Collapse
|
29
|
Peripheral natural killer cell and allo-stimulated T-cell function in kidney transplant recipients associate with cancer risk and immunosuppression-related complications. Kidney Int 2015; 88:1374-1382. [PMID: 26266834 DOI: 10.1038/ki.2015.237] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/05/2015] [Accepted: 06/12/2015] [Indexed: 12/12/2022]
Abstract
Reducing immunosuppression has been proposed as a means of preventing cancer in kidney transplant recipients but this can precipitate graft rejection. Here we tested whether anti-tumor natural killer (NK) cell and allo-responsive T-cell function in kidney transplant recipients may predict cancer risk and define risk of rejection. NK cell function was measured by the release of lactate dehydrogenase and T-cell allo-response by interferon-γ quantification using a panel of reactive T-cell enzyme-linked immunospot (ELISPOT) in 56 kidney transplant recipients with current or past cancer and 26 kidney transplant recipients without cancer. NK function was significantly impaired and the allo-response was significantly lower in kidney transplant recipients with cancer. With prospective follow-up, kidney transplant recipients with poor NK cell function had a hazard ratio of 2.1 (95% confidence interval 0.97-5.00) for the combined end point of metastatic cancer, cancer-related death, or septic death. Kidney transplant recipients with low interferon-γ release were also more likely to reach this combined end point. Thus, posttransplant monitoring of allo-immunity and NK cell function is useful for assessing the risk of over immunosuppression for the development of malignancy and/or death from cancer or sepsis.
Collapse
|
30
|
CD27low natural killer cells prolong allograft survival in mice by controlling alloreactive CD8+ T cells in a T-bet-dependent manner. Transplantation 2015; 99:391-9. [PMID: 25606781 DOI: 10.1097/tp.0000000000000585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Natural killer (NK) cells play a dichotomous role in alloimmune responses because they are known to promote both allograft survival and rejection. The aim of this study was to investigate the role of functionally distinct NK cell subsets in alloimmunity with the hypothesis that this dichotomy is explained by the functional heterogeneity of distinct NK cell subsets. METHODS Because T-bet controls thematuration of NK cells from CD27high to terminally differentiated CD27low NK cells, we used Rag−/−T-bet−/− mice that lackmature CD27low NK cells to study the distinct roles of CD27low versus CD27high NK cells in a model of Tcell–mediated skin transplant rejection under costimulatory blockade conditions. RESULTS We found that T cell–reconstituted Rag1−/− recipients (possessing CD27low NK cells) show significantly prolonged allograft survival on costimulatory blockade when compared to Rag1−/−T-bet−/− mice (lacking CD27low NK cells), indicating that CD27low but not CD27high NK cells enhance allograft survival. Critically, Rag1−/−T-bet−/− recipients showed strikingly increased alloreactive memory CD8+ Tcell responses, as indicated by increased CD8+ Tcell proliferation and interferon-γ production. Therefore, we speculated that CD27low NK cells directly regulate alloreactive CD8+ Tcell responses under costimulatory blockade conditions. To test this, we adoptively transferred CD27low NK cells into Rag1−/−T-bet−/− skin transplant recipients and found that the CD27low NK cells restore better allograft survival by inhibiting the proliferation of alloreactive interferon-γ+CD8+ T cells. CONCLUSIONS In summary, mature CD27low NK cells promote allograft survival under costimulatory blockade conditions by regulating alloreactive memory CD8+ T-cell responses.
Collapse
|
31
|
Weinger JG, Plaisted WC, Maciejewski SM, Lanier LL, Walsh CM, Lane TE. Activating receptor NKG2D targets RAE-1-expressing allogeneic neural precursor cells in a viral model of multiple sclerosis. Stem Cells 2015; 32:2690-701. [PMID: 24898518 DOI: 10.1002/stem.1760] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 04/25/2014] [Accepted: 04/25/2014] [Indexed: 12/11/2022]
Abstract
Transplantation of major histocompatibility complex-mismatched mouse neural precursor cells (NPCs) into mice persistently infected with the neurotropic JHM strain of mouse hepatitis virus (JHMV) results in rapid rejection that is mediated, in part, by T cells. However, the contribution of the innate immune response to allograft rejection in a model of viral-induced neurological disease has not been well defined. Herein, we demonstrate that the natural killer (NK) cell-expressing-activating receptor NKG2D participates in transplanted allogeneic NPC rejection in mice persistently infected with JHMV. Cultured NPCs derived from C57BL/6 (H-2(b) ) mice express the NKG2D ligand retinoic acid early precursor transcript (RAE)-1 but expression was dramatically reduced upon differentiation into either glia or neurons. RAE-1(+) NPCs were susceptible to NK cell-mediated killing whereas RAE-1(-) cells were resistant to lysis. Transplantation of C57BL/6-derived NPCs into JHMV-infected BALB/c (H-2(d) ) mice resulted in infiltration of NKG2D(+) CD49b(+) NK cells and treatment with blocking antibody specific for NKG2D increased survival of allogeneic NPCs. Furthermore, transplantation of differentiated RAE-1(-) allogeneic NPCs into JHMV-infected BALB/c mice resulted in enhanced survival, highlighting a role for the NKG2D/RAE-1 signaling axis in allograft rejection. We also demonstrate that transplantation of allogeneic NPCs into JHMV-infected mice resulted in infection of the transplanted cells suggesting that these cells may be targets for infection. Viral infection of cultured cells increased RAE-1 expression, resulting in enhanced NK cell-mediated killing through NKG2D recognition. Collectively, these results show that in a viral-induced demyelination model, NK cells contribute to rejection of allogeneic NPCs through an NKG2D signaling pathway.
Collapse
Affiliation(s)
- Jason G Weinger
- Department of Molecular Biology & Biochemistry; Sue and Bill Gross Stem Cell Center, University of California, Irvine, California, USA
| | | | | | | | | | | |
Collapse
|
32
|
Chong AS, Alegre ML. Transplantation tolerance and its outcome during infections and inflammation. Immunol Rev 2015; 258:80-101. [PMID: 24517427 DOI: 10.1111/imr.12147] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Much progress has been made toward understanding the mechanistic basis of transplantation tolerance in experimental models, which implicates clonal deletion of alloreactive T and B cells, induction of cell-intrinsic hyporesponsiveness, and dominant regulatory cells mediating infectious tolerance and linked suppression. Despite encouraging success in the laboratory, achieving tolerance in the clinic remains challenging, although the basis for these challenges is beginning to be understood. Heterologous memory alloreactive T cells generated by infections prior to transplantation have been shown to be a critical barrier to tolerance induction. Furthermore, infections at the time of transplantation and tolerance induction provide a pro-inflammatory milieu that alters the stability and function of regulatory T cells as well as the activation requirements and differentiation of effector T cells. Thus, infections can result in enhanced alloreactivity, resistance to tolerance induction, and destabilization of the established tolerance state. We speculate that these experimental findings have relevance to the clinic, where infections have been associated with allograft rejection and may be a causal event precipitating the loss of grafts after long periods of stable operational tolerance. Understanding the mechanisms by which infections prevent and destabilize tolerance can lead to therapies that promote stable life-long tolerance in transplant recipients.
Collapse
Affiliation(s)
- Anita S Chong
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
33
|
Dai H, Peng F, Lin M, Xia J, Yu S, Lan G, Wang Y, Xie X, Fang C, Corbascio M, Qi Z, Peng L. Anti-OX40L monoclonal antibody prolongs secondary heart allograft survival based on CD40/CD40L and LFA-1/ICAM-1 blockade. Transpl Immunol 2015; 32:84-91. [PMID: 25613092 DOI: 10.1016/j.trim.2015.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/11/2015] [Accepted: 01/12/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Memory T cells (Tms) form a barrier against long-term allograft survival; however, CD4(+)Foxp3(+) regulatory T cells (Tregs) can suppress allograft rejection. The OX40/OX40L pathway is critical to the generation of Tms and turns off Treg suppressor function. METHODS B6 mice that rejected BALB/c skin grafts after 4 weeks were used as the secondary heart transplant recipients. The skin recipient mice, termed S0, S2 and S3, were treated with the isotype antibodies, anti-CD40L/LFA-1 or anti-OX40L combined with anti-CD40L/LFA-1 mAbs, respectively. The secondary heart recipients, termed H0 and H2, received anti-CD40L/LFA-1 mAbs or not, respectively (Fig. 1). RESULTS Four weeks after primary skin transplantation, the Tms in the S3 group that received anti-OX40L with anti-CD40L/LFA-1 mAbs were reduced compared to those in the S2 group (CD4(+) Tm: 32.61 ± 2.20% in S2 vs. 25.36 ± 1.16% in S3; CD8(+) Tm: 27.76 ± 1.96% in S2 vs. 20.95 ± 1.30% in S3; P < 0.01). Meanwhile, the proportions of Tregs in S3 increased compared to those in S2 (P < 0.05). The anti-OX40L with anti-CD40L/LFA-1 mAbs group (S3H2) prolonged the mean survival time (MST) following secondary heart transplantation from 9.5 days to 21 days (P < 0.001). Furthermore, allogeneic proliferation of recipient splenic T cells and graft-infiltrating lymphocytes were significantly inhibited in the S3H2 group. Additionally, a higher level of IL-10 was detected in sera and allografts. CONCLUSIONS Anti-OX40L mAb could prolong secondary heart allograft survival based on CD40/CD40L and LFA-1/ICAM-1 blockade. The mechanism of protecting allografts using anti-OX40L mAb involved impairing the generation of Tm and up-regulating IL-10 producing Tregs, inhibiting the function of T cells.
Collapse
Affiliation(s)
- Helong Dai
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Fenghua Peng
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Minjie Lin
- Department of Cardiology, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Junjie Xia
- Organ Transplantation Institute, Xiamen University, Fujian Province, PR China
| | - Shaojie Yu
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Gongbin Lan
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Yu Wang
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Xubiao Xie
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | - Chunhua Fang
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China
| | | | - Zhongquan Qi
- Organ Transplantation Institute, Xiamen University, Fujian Province, PR China
| | - Longkai Peng
- Department of Urological Organ Transplantation, Center of Organ Transplantation, Second Xiangya Hospital, Central South University, Hunan Province, PR China.
| |
Collapse
|
34
|
Abstract
Natural killer (NK) cells are effector cells of the innate immune system that can lyse target cells without prior sensitization and have an important role in host defense to pathogens and transformed cells. A balance between negative and positive signals transmitted via germ line-encoded inhibitory and activating receptors controls the function of NK cells. Although the concept of "missing-self" would suggest that NK cells could target foreign allografts, the prevailing dogma has been that NK cells are not active participants in the mechanisms that culminate in the rejection of solid organ allografts. Recent studies, however, challenge this conclusion and instead implicate NK cells in contributing to both graft rejection and tolerance to an allograft. In this review, we highlight recent studies with the goal of understanding the complex NK cell interactions that impact alloimmunity.
Collapse
Affiliation(s)
- Uzi Hadad
- Division of Abdominal Transplantation, Department of Surgery and Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | |
Collapse
|
35
|
Pawlick R, Gala-Lopez B, Pepper AR, McCall M, Ziff O, Shapiro AMJ. The combination of anti-NKG2D and CTLA-4 Ig therapy prolongs islet allograft survival in a murine model. Am J Transplant 2014; 14:2367-74. [PMID: 25179027 DOI: 10.1111/ajt.12838] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 05/05/2014] [Accepted: 05/08/2014] [Indexed: 01/25/2023]
Abstract
Islet transplantation is an effective means of treating severe type 1 diabetes in patients with life-threatening hypoglycemia. Improvements in glycemic control with correction of HbA1C enhance quality of life irrespective of insulin independence. By antagonizing the Natural Killer Group 2, member D (NKG2D) receptor expression on NK and CD8+ T cells, in combination with blocking CTLA-4 binding sites, we demonstrate a significant delay of graft rejection in islet allotransplant. Anti-NKG2D combined with CTLA-4 Ig (n = 15) results in prolonged allograft survival, with 84.6 ± 10% of the recipients displaying insulin independence compared to controls (n = 10, p < 0.001). The effect of combination therapy on graft survival is superior to treatments alone (CTLA-4 Ig vs. combination p = 0.024, anti-NKG2D vs. combination p < 0.001) indicating an interaction between these pathways. In addition, combination treatment also improves glucose tolerance when compared to controls (n = 10, p = 0.018). Histologically, NKG2D+ cells were significantly decreased within the allograft after 7 days of combination treatment (n = 6, p = 0.029). T cell proliferation was significantly reduced with anti-NKG2D therapy and CD8+ T cell daughter fractions were also significantly decreased with mAb and combination treatment when measured by in vitro mixed lymphocyte reaction (n = 5, p = 0.015, p = 0.005 and p = 0.048). These results demonstrate that inhibition of NKG2D receptors and costimulatory pathways enhance islet allograft survival.
Collapse
Affiliation(s)
- R Pawlick
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | | | |
Collapse
|
36
|
Krupnick AS, Lin X, Li W, Higashikubo R, Zinselmeyer BH, Hartzler H, Toth K, Ritter JH, Berezin MY, Wang ST, Miller MJ, Gelman AE, Kreisel D. Central memory CD8+ T lymphocytes mediate lung allograft acceptance. J Clin Invest 2014; 124:1130-43. [PMID: 24569377 PMCID: PMC3938255 DOI: 10.1172/jci71359] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 12/05/2013] [Indexed: 12/31/2022] Open
Abstract
Memory T lymphocytes are commonly viewed as a major barrier for long-term survival of organ allografts and are thought to accelerate rejection responses due to their rapid infiltration into allografts, low threshold for activation, and ability to produce inflammatory mediators. Because memory T cells are usually associated with rejection, preclinical protocols have been developed to target this population in transplant recipients. Here, using a murine model, we found that costimulatory blockade-mediated lung allograft acceptance depended on the rapid infiltration of the graft by central memory CD8+ T cells (CD44(hi)CD62L(hi)CCR7+). Chemokine receptor signaling and alloantigen recognition were required for trafficking of these memory T cells to lung allografts. Intravital 2-photon imaging revealed that CCR7 expression on CD8+ T cells was critical for formation of stable synapses with antigen-presenting cells, resulting in IFN-γ production, which induced NO and downregulated alloimmune responses. Thus, we describe a critical role for CD8+ central memory T cells in lung allograft acceptance and highlight the need for tailored approaches for tolerance induction in the lung.
Collapse
Affiliation(s)
- Alexander Sasha Krupnick
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Xue Lin
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Wenjun Li
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ryuiji Higashikubo
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bernd H. Zinselmeyer
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Hollyce Hartzler
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kelsey Toth
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jon H. Ritter
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mikhail Y. Berezin
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Steven T. Wang
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mark J. Miller
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Andrew E. Gelman
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Daniel Kreisel
- Department of Surgery and
Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Department of Radiology and
Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
37
|
Abstract
Organ transplantation appears today to be the best alternative to replace the loss of vital organs induced by various diseases. Transplants can, however, also be rejected by the recipient. In this review, we provide an overview of the mechanisms and the cells/molecules involved in acute and chronic rejections. T cells and B cells mainly control the antigen-specific rejection and act either as effector, regulatory, or memory cells. On the other hand, nonspecific cells such as endothelial cells, NK cells, macrophages, or polymorphonuclear cells are also crucial actors of transplant rejection. Last, beyond cells, the high contribution of antibodies, chemokines, and complement molecules in graft rejection is discussed in this article. The understanding of the different components involved in graft rejection is essential as some of them are used in the clinic as biomarkers to detect and quantify the level of rejection.
Collapse
Affiliation(s)
- Aurélie Moreau
- INSERM UMR 1064, Center for Research in Transplantation and Immunology-ITUN, CHU de Nantes 44093, France
| | | | | | | |
Collapse
|
38
|
Jungraithmayr W, Codarri L, Bouchaud G, Krieg C, Boyman O, Gyülvészi G, Becher B, Weder W, Münz C. Cytokine complex-expanded natural killer cells improve allogeneic lung transplant function via depletion of donor dendritic cells. Am J Respir Crit Care Med 2013; 187:1349-59. [PMID: 23590269 DOI: 10.1164/rccm.201209-1749oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Natural killer (NK) cells are innate lymphocytes that target virus-infected and tumor cells. Much less is known about their ability to limit adaptive immune responses. OBJECTIVES Thus, we investigated to what extent NK cells can influence mouse lung allograft rejection. METHODS For this purpose, we employed an orthotopic lung transplantation model in mice. MEASUREMENTS AND MAIN RESULTS We demonstrate here that NK cells infiltrate mouse lung allografts before T cells and thereby diminished allograft inflammation, and that NK-cell deficiency enhanced allograft rejection. In contrast, expansion of recipient NK cells through IL-15/IL-15Rα complex treatment resulted in decreased T-cell infiltration and alloreactive T-cell priming as well as improved function of the allogeneic lung transplant. Only perforin-competent, but not perforin-deficient, NK cells were able to transfer these beneficial effects into transplanted NK cell-deficient IL-15Rα(-/-) mice. These NK cells killed allogeneic dendritic cells (DCs) in vitro and significantly decreased the number of allogeneic DCs in transplanted lungs in vivo. Furthermore, DC-depleted lung allografts presented decreased signs of rejection. CONCLUSIONS These results suggest that NK cells favor allograft acceptance by depleting donor-derived DCs, which otherwise would prime alloreactive T-cell responses. Thus, conditioning regimens that augment NK-cell reactivity should be clinically explored to prepare lung allograft recipients.
Collapse
Affiliation(s)
- Wolfgang Jungraithmayr
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Smelt MJ, Faas MM, de Haan BJ, de Haan A, Vaage JT, de Vos P. The role of alloresponsive Ly49+ NK cells in rat islet allograft failure in the presence and absence of cytomegalovirus. Cell Transplant 2013; 23:1381-94. [PMID: 23866824 DOI: 10.3727/096368913x670930] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
There are still many factors to discover to explain the low success rates of islet allografts. In this study, we demonstrate that specific subpopulations of alloreactive NK cells may be involved in the failure of islet allografts. By performing allotransplantation in rats (n = 13), we observed peripheral expansion and infiltration of alloreactive Ly49i2(+) NK cells in the grafts. An effective strategy in rats to enhance the expansion of Ly49i2(+) NK cells is performing a rat cytomegalovirus infection (n = 6). Cytomegalovirus infection was associated with an early expansion of the Ly49i2(+) NK cells and accelerated islet graft failure. The Ly49i2(+) NK cells are both alloreactive and involved in virus clearance. The expansion of this subpopulation could not be blocked by cyclosporin A immunosuppression. Also alloreactive KLRH1(+) NK cells infiltrated the grafts, but nonalloreactive NKR-P1B(+) cells were not observed in the islet allografts. Perforin staining of the infiltrating NK cells demonstrated the cytotoxic capacity of these cells. Our data suggest a role for this NK subpopulation in rat islet allograft destruction.
Collapse
Affiliation(s)
- Maaike J Smelt
- Department of Pathology and Medical Biology, Division of Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW A series of recent studies defy conventional wisdom by showing that natural killer (NK) cells exert a powerful and long-lasting influence on the immune response to whole organ allografts. The early activation of NK cells following transplantation is associated with killing of allogeneic target cells and release of immunomodulatory chemokines and cytokines, which can contribute to either rejection or tolerance. Here, we review findings describing NK cell receptors, potential mediators and mechanisms underlying the dual influence of NK cells in solid organ transplantation. RECENT FINDINGS New studies show that NK cells can discriminate between self and foreign tissues and play a key role in the initiation and regulation of adaptive immune responses after solid organ transplantation. Depending upon the types of NK cell receptors engaged and the nature of cytokines released, early NK cell activation can promote either rejection or tolerance. SUMMARY Solid organ transplantation is associated with the early activation of NK cells, which are then licensed to kill allogeneic target cells directly or via antibody-dependent cellular cytotoxicity and release various chemokines and immunomodulatory cytokines. Depending upon the nature of NK cell subsets activated and their ability to kill allogeneic target cells and release certain types of cytokines, NK cells can promote the activation/expansion of pro-inflammatory Th1 cells or regulatory Th2/Treg cells thus tilting the balance of alloimmunity towards rejection or tolerance. An in-depth understanding of these mechanisms will be necessary in order to design therapies targeting NK cells in human transplantation.
Collapse
|
41
|
Crome SQ, Lang PA, Lang KS, Ohashi PS. Natural killer cells regulate diverse T cell responses. Trends Immunol 2013; 34:342-9. [PMID: 23601842 DOI: 10.1016/j.it.2013.03.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 03/13/2013] [Accepted: 03/14/2013] [Indexed: 12/15/2022]
Abstract
Natural killer (NK) cells are important mediators of the immune response against microbial pathogens and tumors. There is growing evidence from mouse and human studies that, NK cells exhibit immunoregulatory functions and can limit T cell immunity. NK cell regulatory activity has been demonstrated in a variety of disease models including chronic viral infection, autoimmunity, and transplantation. Depending on the nature of the immune challenge, NK cells use different strategies to limit T cell function, including via cytokines, interactions with NK receptors NKG2D and NKp46, or by perforin-mediated T cell death. Future work should address whether specific subsets of NK cells inhibit T cell responses, and how NK cells acquire immunosuppressive functions.
Collapse
Affiliation(s)
- Sarah Q Crome
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | | | | | | |
Collapse
|
42
|
D’Addio F, Ueno T, Clarkson M, Zhu B, Vergani A, Freeman GJ, Sayegh MH, Ansari MJI, Fiorina P, Habicht A. CD160Ig fusion protein targets a novel costimulatory pathway and prolongs allograft survival. PLoS One 2013; 8:e60391. [PMID: 23593209 PMCID: PMC3617215 DOI: 10.1371/journal.pone.0060391] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/26/2013] [Indexed: 12/30/2022] Open
Abstract
CD160 is a cell surface molecule expressed by most NK cells and approximately 50% of CD8(+) cytotoxic T lymphocytes. Engagement of CD160 by MHC class-I directly triggers a costimulatory signal to TCR-induced proliferation, cytokine production and cytotoxic effector functions. The role of CD160 in alloimmunity is unknown. Using a newly generated CD160 fusion protein (CD160Ig) we examined the role of the novel costimulatory molecule CD160 in mediating CD4(+) or CD8(+) T cell driven allograft rejection. CD160Ig inhibits alloreactive CD8(+) T cell proliferation and IFN-γ production in vitro, in particular in the absence of CD28 costimulation. Consequently CD160Ig prolongs fully mismatched cardiac allograft survival in CD4(-/-), CD28(-/-) knockout and CTLA4Ig treated WT recipients, but not in WT or CD8(-/-) knockout recipients. The prolonged cardiac allograft survival is associated with reduced alloreactive CD8(+) T cell proliferation, effector/memory responses and alloreactive IFN-γ production. Thus, CD160 signaling is particularly important in CD28-independent effector/memory CD8(+) alloreactive T cell activation in vivo and therefore may serve as a novel target for prevention of allograft rejection.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, CD/genetics
- Antigens, CD/immunology
- CD28 Antigens/deficiency
- CD28 Antigens/immunology
- CD4 Antigens/genetics
- CD4 Antigens/immunology
- Cytokines/biosynthesis
- Cytotoxicity, Immunologic/genetics
- Cytotoxicity, Immunologic/immunology
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/immunology
- Gene Expression
- Graft Survival/drug effects
- Graft Survival/genetics
- Graft Survival/immunology
- Heart Transplantation/immunology
- Heart Transplantation/mortality
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/immunology
- Immunoglobulin G/genetics
- Immunoglobulin G/immunology
- Immunologic Memory/genetics
- Immunologic Memory/immunology
- Interferon-gamma/biosynthesis
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacology
- Signal Transduction/drug effects
- Skin Transplantation/immunology
- Skin Transplantation/mortality
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Transplantation, Homologous
Collapse
Affiliation(s)
- Francesca D’Addio
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- Transplantation and Internal Medicine Division, San Raffaele Scientific Institute, Milan, Italy
| | - Takuya Ueno
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael Clarkson
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Renal Medicine, Cork University Hospital, Cork, Ireland
| | - Baogong Zhu
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Andrea Vergani
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Mohamed H. Sayegh
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mohammed Javeed I. Ansari
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paolo Fiorina
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- Transplantation and Internal Medicine Division, San Raffaele Scientific Institute, Milan, Italy
| | - Antje Habicht
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- Transplant Center Munich - LMU, University Hospital, Munich, Germany
| |
Collapse
|
43
|
The innate natural killer cells in transplant rejection and tolerance induction. Curr Opin Organ Transplant 2012; 13:339-43. [PMID: 18685327 DOI: 10.1097/mot.0b013e3283061115] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW The roles of adaptive immune cells in transplant models have been extensively studied, but very little is known about the role of innate immune cells in the allograft response, especially in tolerance induction. In this review, we summarized the latest developments in the study of the role of natural killer cells in mediating graft rejection and tolerance induction. RECENT FINDINGS Natural killer cells are potent cytolytic cells; they also induce tissue inflammation by producing powerful proinflammatory cytokines. Thus, natural killer cells can act as effector cells in transplant rejection. Recent studies, however, have demonstrated additional roles for natural killer cells in the induction of transplant tolerance. We found that natural killer cells control survival of graft-derived donor cells and killing of donor dendritic cells by host natural killer cells inhibits direct priming of alloreactive T cells. Natural killer cells are also shown directly to suppress the activation of T cells. In other models, natural killer cells are found to regulate the induction of regulatory T cells. These new findings may have important clinical implications in tolerance induction. SUMMARY Natural killer cells are involved in both graft rejection and tolerance induction; such opposing effects may be mediated by differences in the activation status of natural killer cells. We believe that natural killer cells can be therapeutically modified for the induction of transplant tolerance.
Collapse
|
44
|
Lee B, Oh CK, Kim MS, Kim JH, Kim SJ, Kim HS, Shin GT. Cytokine gene expression in peripheral blood mononuclear cells during acute renal allograft rejection. Transplant Proc 2012; 44:236-40. [PMID: 22310622 DOI: 10.1016/j.transproceed.2011.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Many studies have explored the participation of cytokines and their genes in renal allograft rejection by using biopsy tissues. To screen for rejection, a biopsy is too invasive to perform without a clinical clue. Therefore, we studied the expression of cytokines that contribute to the early phase of allograft rejection by analyzing mRNA transcripts in sequential blood samples of peripheral blood mononuclear cells (PBMCs) 120 of 6 among patients transplanted before diagnosis of rejection. for comparison with 6 control recipients. The relative expression amount of cytokine genes encoding interleukin (IL) 2, IL-4, IL-10, IL-15, and interferon-γ were assessed using real-time reverse-transcription polymerase chain reactions. IL-2, IL-4, and IL-15 mRNA expressions in clinically stable prerejection phase of the rejection group were significantly higher than those of the control group. In the prerejection samples, the expression of mRNA encoding IL-10 negatively correlated with the expressions of IL-2, IL-4, and IL-15 mRNAs, which were not different from the positive correlations in the postoperative samples from the control group. The expression patterns of IL-2, IL-4, IL-10, and IL-15 genes in PBMCs after transplantation may help to identify acute rejection episodes before clinical deterioration to monitor the efficacy of immunosuppressive treatment.
Collapse
Affiliation(s)
- B Lee
- Department of Surgery, Seoul Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
45
|
Benichou G, Tonsho M, Tocco G, Nadazdin O, Madsen JC. Innate immunity and resistance to tolerogenesis in allotransplantation. Front Immunol 2012; 3:73. [PMID: 22566954 PMCID: PMC3342343 DOI: 10.3389/fimmu.2012.00073] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/22/2012] [Indexed: 01/06/2023] Open
Abstract
The development of immunosuppressive drugs to control adaptive immune responses has led to the success of transplantation as a therapy for end-stage organ failure. However, these agents are largely ineffective in suppressing components of the innate immune system. This distinction has gained in clinical significance as mounting evidence now indicates that innate immune responses play important roles in the acute and chronic rejection of whole organ allografts. For instance, whereas clinical interest in natural killer (NK) cells was once largely confined to the field of bone marrow transplantation, recent findings suggest that these cells can also participate in the acute rejection of cardiac allografts and prevent tolerance induction. Stimulation of Toll-like receptors (TLRs), another important component of innate immunity, by endogenous ligands released in response to ischemia/reperfusion is now known to cause an inflammatory milieu favorable to graft rejection and abrogation of tolerance. Emerging data suggest that activation of complement is linked to acute rejection and interferes with tolerance. In summary, the conventional wisdom that the innate immune system is of little importance in whole organ transplantation is no longer tenable. The addition of strategies that target TLRs, NK cells, complement, and other components of the innate immune system will be necessary to eventually achieve long-term tolerance to human allograft recipients.
Collapse
Affiliation(s)
- Gilles Benichou
- Transplant Research Center, Massachusetts General Hospital and Harvard Medical School Boston, MA, USA
| | | | | | | | | |
Collapse
|
46
|
Conlon TM, Saeb-Parsy K, Cole JL, Motallebzadeh R, Qureshi MS, Rehakova S, Negus MC, Callaghan CJ, Bolton EM, Bradley JA, Pettigrew GJ. Germinal center alloantibody responses are mediated exclusively by indirect-pathway CD4 T follicular helper cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:2643-52. [PMID: 22323543 DOI: 10.4049/jimmunol.1102830] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The durable alloantibody responses that develop in organ transplant patients indicate long-lived plasma cell output from T-dependent germinal centers (GCs), but which of the two pathways of CD4 T cell allorecognition is responsible for generating allospecific T follicular helper cells remains unclear. This was addressed by reconstituting T cell-deficient mice with monoclonal populations of TCR-transgenic CD4 T cells that recognized alloantigen only as conformationally intact protein (direct pathway) or only as self-restricted allopeptide (indirect pathway) and then assessing the alloantibody response to a heart graft. Recipients reconstituted with indirect-pathway CD4 T cells developed long-lasting IgG alloantibody responses, with splenic GCs and allospecific bone marrow plasma cells readily detectable 50 d after heart transplantation. Differentiation of the transferred CD4 T cells into T follicular helper cells was confirmed by follicular localization and by acquisition of signature phenotype. In contrast, IgG alloantibody was not detectable in recipient mice reconstituted with direct-pathway CD4 T cells. Neither prolongation of the response by preventing NK cell killing of donor dendritic cells nor prior immunization to develop CD4 T cell memory altered the inability of the direct pathway to provide allospecific B cell help. CD4 T cell help for GC alloantibody responses is provided exclusively via the indirect-allorecognition pathway.
Collapse
Affiliation(s)
- Thomas M Conlon
- Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Zecher D, Li Q, Williams AL, Walters JT, Baddoura FK, Chalasani G, Rothstein DM, Shlomchik WD, Demetris AJ, Lakkis FG. Innate immunity alone is not sufficient for chronic rejection but predisposes healed allografts to T cell-mediated pathology. Transpl Immunol 2011; 26:113-8. [PMID: 22226941 DOI: 10.1016/j.trim.2011.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 12/13/2011] [Accepted: 12/13/2011] [Indexed: 11/24/2022]
Abstract
BACKGROUND Acute allograft rejection is dependent on adaptive immunity, but it is unclear whether the same is true for chronic rejection. Here we asked whether innate immunity alone is sufficient for causing chronic rejection of mouse cardiac allografts. METHODS We transplanted primarily vascularized cardiac grafts to recombinase activating gene-knockout (RAG(-/-)) mice that lack T and B cells but have an intact innate immune system. Recipients were left unmanipulated, received adjuvants that stimulate innate immunity, or were reconstituted with B-1 lymphocytes to generate natural IgM antibodies. In a second model, we transplanted cardiac allografts to mice that lack secondary lymphoid tissues (splenectomized aly/aly recipients) and studied the effect of NK cell inactivation on T cell-mediated chronic rejection. RESULTS Acute cardiac allograft rejection was not observed in any of the recipients. Histological analysis of allografts harvested 50 to 90 days after transplantation to RAG(-/-) mice failed to identify chronic vascular or parenchymal changes beyond those observed in control syngeneic grafts. Chronic rejection of cardiac allografts parked in splenectomized aly/aly mice was observed only after the transfer of exogenously activated T cells. NK inactivation throughout the experiment, or during the parking period alone, reduced the severity of T cell-dependent chronic rejection. CONCLUSIONS The innate immune system alone is not sufficient for causing chronic rejection. NK cells predispose healed allografts to T cell-dependent chronic rejection and may contribute to chronic allograft pathology.
Collapse
Affiliation(s)
- Daniel Zecher
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Salvadori M. What is the purpose of launching the World Journal of Transplantation? World J Transplant 2011; 1:1-3. [PMID: 24175186 PMCID: PMC3782228 DOI: 10.5500/wjt.v1.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 12/07/2011] [Accepted: 12/19/2011] [Indexed: 02/05/2023] Open
Abstract
Congratulations to the publisher, members of the editorial board of the journal, all the authors and readers for launching the World Journal of Transplantation (WJT) as a new member of the World series journal family. Transplantations are rapidly evolving and share knowledge with a number of basic and clinical sciences: molecular biology, stem cell investigators, immune system, pharmacology, biotechnology, surgery and physicians of different organs such as the kidneys, liver, heart, lung, bone marrow and so on. The WJT is a peer reviewed open access journal centered on the different fields involved in transplant activity. If you want to share your experiences and new findings in the field of transplantation with your peers you will find the WJT a good media to publish your papers.
Collapse
Affiliation(s)
- Maurizio Salvadori
- Maurizio Salvadori, Renal Unit, Department of Renal Transplantation, Careggi University Hospital, viale Pieraccini 18, 50139 Florence, Italy
| |
Collapse
|
49
|
Karp SJ, Mannon RB. What's New, What's Hot in Solid Organ Transplantation? Summary of the American Transplant Congress 2011. Am J Transplant 2011; 11:2308-16. [PMID: 21967066 DOI: 10.1111/j.1600-6143.2011.03774.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Breakthroughs in basic and clinical science in solid organ transplantation were presented at the American Transplant Congress 2011. Key areas of presentation included the pathogenesis of late allograft failure, immune regulation and tolerance, pathways in allograft injury, electing appropriate patients for transplantation, determining the best allocation schemes to maximize effective utilization, organ preservation, monitoring the alloimmune response and immunosuppressive management. In this review, we present highlights of the meeting. These presentations demonstrate the exciting promise in translating from the bench to affect patient care.
Collapse
Affiliation(s)
- S J Karp
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | |
Collapse
|
50
|
Suárez-Álvarez B, Fernández-Sánchez A, López-Vázquez A, Coto E, Ortega F, López-Larrea C. NKG2D and its ligands: active factors in the outcome of solid organ transplantation? Kidney Int Suppl (2011) 2011; 1:52-57. [PMID: 25018903 PMCID: PMC4089716 DOI: 10.1038/kisup.2011.13] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The role of natural killer (NK) cells in solid organ transplantation is not well established, although several recent reports highlight the importance of the activating receptor NKG2D and its ligands in the development of rejection during transplantation. The human NKG2D ligands (MICA and MICB) are induced in allografts during acute and chronic rejection, and the presence of anti-MICA antibodies is correlated with a higher incidence of rejection. The binding of these ligands to its receptor NKG2D activates NK cells, enhances the functions of effectors, and allows NK cells to function as a bridge between innate and adaptive immunity associated with the transplantation. In fact, blockage of NKG2D with the anti-NKG2D monoclonal antibodies prolongs graft survival and prevents CD28-independent rejection in heart and skin allograft mouse models. Furthermore, the current immunosuppressive therapies can modulate the expression of NK cell receptors and consequently the effector functions of NK cells. That is particularly important during the first few months after transplantation, when the susceptibility to opportunistic viral infections is higher and NKG2D has an essential role. In this review, we analyze in detail the potential role of the NKG2D-activating receptor and its ligands in the immune responses during the outcome of solid organ transplantation. These findings open a new pathway for therapeutic intervention that can contribute to tolerance in solid organ transplantation.
Collapse
Affiliation(s)
| | | | - Antonio López-Vázquez
- Department of Immunology, Hospital Universitario Central de Asturias , Oviedo, Spain
| | - Eliecer Coto
- Department of Molecular Genetics, Hospital Universitario Central de Asturias , Oviedo, Spain ; Fundación Renal 'Iñigo Alvarez de Toledo' , Madrid, Spain
| | - Francisco Ortega
- Fundación Renal 'Iñigo Alvarez de Toledo' , Madrid, Spain ; Department of Nephrology, Hospital Universitario Central de Asturias , Oviedo, Spain
| | - Carlos López-Larrea
- Department of Immunology, Hospital Universitario Central de Asturias , Oviedo, Spain ; Fundación Renal 'Iñigo Alvarez de Toledo' , Madrid, Spain
| |
Collapse
|