1
|
Gill RG. Bringing Clarity to the Murky Problem of Cardiac Allograft Vasculopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:986-989. [PMID: 35577009 PMCID: PMC9253909 DOI: 10.1016/j.ajpath.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Ronald G Gill
- Department of Surgery, University of Colorado Denver, Aurora, Colorado.
| |
Collapse
|
2
|
Tsuda H, Dvorina N, Keslar KS, Nevarez-Mejia J, Valenzuela NM, Reed EF, Fairchild RL, Baldwin WM. Molecular Signature of Antibody-Mediated Chronic Vasculopathy in Heart Allografts in a Novel Mouse Model. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1053-1065. [PMID: 35490714 PMCID: PMC9253905 DOI: 10.1016/j.ajpath.2022.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 04/23/2023]
Abstract
Cardiac allograft vasculopathy (CAV) limits the long-term success of heart transplants. Generation of donor-specific antibodies (DSAs) is associated with increased incidence of CAV clinically, but mechanisms underlying development of this pathology remain poorly understood. Major histocompatibility complex-mismatched A/J cardiac allografts in B6.CCR5-/- recipients have been reported to undergo acute rejection with little T-cell infiltration, but intense deposition of C4d in large vessels and capillaries of the graft accompanied by high titers of DSA. This model was modified to investigate mechanisms of antibody-mediated CAV by transplanting A/J hearts to B6.CCR5-/- CD8-/- mice that were treated with low doses of anti-CD4 monoclonal antibody to decrease T-cell-mediated graft injury and promote antibody-mediated injury. Although the mild inhibition of CD4 T cells extended allograft survival, the grafts developed CAV with intense C4d deposition and macrophage infiltration by 14 days after transplantation. Development of CAV correlated with recipient DSA titers. Transcriptomic analysis of microdissected allograft arteries identified the Notch ligand Dll4 as the most elevated transcript in CAV, associated with high versus low titers of DSA. More importantly, these analyses revealed a differential expression of transcripts in the CAV lesions compared with the matched apical tissue that lacks large arteries. In conclusion, these findings report a novel model of antibody-mediated CAV with the potential to facilitate further understanding of the molecular mechanisms promoting development of CAV.
Collapse
Affiliation(s)
- Hidetoshi Tsuda
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Nina Dvorina
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Karen S Keslar
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jessica Nevarez-Mejia
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Nicole M Valenzuela
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Elaine F Reed
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Robert L Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| | - William M Baldwin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
3
|
Borges TJ, Murakami N, Lape IT, Gassen RB, Liu K, Cai S, Daccache J, Safa K, Shimizu T, Ohori S, Paterson AM, Cravedi P, Azzi J, Sage P, Sharpe A, Li XC, Riella LV. Overexpression of PD-1 on T cells promotes tolerance in cardiac transplantation via an ICOS-dependent mechanism. JCI Insight 2021; 6:142909. [PMID: 34752418 PMCID: PMC8783692 DOI: 10.1172/jci.insight.142909] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/04/2021] [Indexed: 12/04/2022] Open
Abstract
The programmed death 1/programmed death ligand 1 (PD-1/PD-L1) pathway is a potent inhibitory pathway involved in immune regulation and is a potential therapeutic target in transplantation. In this study, we show that overexpression of PD-1 on T cells (PD-1 Tg) promotes allograft tolerance in a fully MHC-mismatched cardiac transplant model when combined with costimulation blockade with CTLA-4–Ig. PD-1 overexpression on T cells also protected against chronic rejection in a single MHC II–mismatched cardiac transplant model, whereas the overexpression still allowed the generation of an effective immune response against an influenza A virus. Notably, Tregs from PD-1 Tg mice were required for tolerance induction and presented greater ICOS expression than those from WT mice. The survival benefit of PD-1 Tg recipients required ICOS signaling and donor PD-L1 expression. These results indicate that modulation of PD-1 expression, in combination with a costimulation blockade, is a promising therapeutic target to promote transplant tolerance.
Collapse
Affiliation(s)
- Thiago J Borges
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, United States of America
| | - Naoka Murakami
- Transplantation Research Center, Brigham & Women's Hospital, Boston, United States of America
| | - Isadora T Lape
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, United States of America
| | - Rodrigo B Gassen
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, United States of America
| | - Kaifeng Liu
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, United States of America
| | - Songjie Cai
- Transplantation Research Center, Brigham & Women's Hospital, Boston, United States of America
| | - Joe Daccache
- Transplantation Research Center, Brigham & Women's Hospital, Boston, United States of America
| | - Kassem Safa
- Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, United States of America
| | - Tetsunosuke Shimizu
- Transplantation Research Center, Brigham & Women's Hospital, Boston, United States of America
| | - Shunsuke Ohori
- Transplantation Research Center, Brigham & Women's Hospital, Boston, United States of America
| | - Alison M Paterson
- Department of Immunobiology, Harvard Medical School, Boston, United States of America
| | - Paolo Cravedi
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Jamil Azzi
- Transplantation Research Center, Brigham & Women's Hospital, Boston, United States of America
| | - Peter Sage
- Transplantation Research Center, Brigham & Women's Hospital, Boston, United States of America
| | - Arlene Sharpe
- Department of Immunology, Harvard Medical School, Boston, United States of America
| | - Xian C Li
- Immunobiology and Transplant Science Center, Houston Methodist Hospital, Houston, United States of America
| | - Leonardo V Riella
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, United States of America
| |
Collapse
|
4
|
Usuelli V, Ben Nasr M, D'Addio F, Liu K, Vergani A, El Essawy B, Yang J, Assi E, Uehara M, Rossi C, Solini A, Capobianco A, Rigamonti E, Potena L, Venturini M, Sabatino M, Bottarelli L, Ammirati E, Frigerio M, Castillo‐Leon E, Maestroni A, Azzoni C, Loretelli C, Joe Seelam A, Tai AK, Pastore I, Becchi G, Corradi D, Visner GA, Zuccotti GV, Chau NB, Abdi R, Pezzolesi MG, Fiorina P. miR-21 antagonism reprograms macrophage metabolism and abrogates chronic allograft vasculopathy. Am J Transplant 2021; 21:3280-3295. [PMID: 33764625 PMCID: PMC8518036 DOI: 10.1111/ajt.16581] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/19/2021] [Accepted: 03/09/2021] [Indexed: 01/25/2023]
Abstract
Despite much progress in improving graft outcome during cardiac transplantation, chronic allograft vasculopathy (CAV) remains an impediment to long-term graft survival. MicroRNAs (miRNAs) emerged as regulators of the immune response. Here, we aimed to examine the miRNA network involved in CAV. miRNA profiling of heart samples obtained from a murine model of CAV and from cardiac-transplanted patients with CAV demonstrated that miR-21 was most significantly expressed and was primarily localized to macrophages. Interestingly, macrophage depletion with clodronate did not significantly prolong allograft survival in mice, while conditional deletion of miR-21 in macrophages or the use of a specific miR-21 antagomir resulted in indefinite cardiac allograft survival and abrogated CAV. The immunophenotype, secretome, ability to phagocytose, migration, and antigen presentation of macrophages were unaffected by miR-21 targeting, while macrophage metabolism was reprogrammed, with a shift toward oxidative phosphorylation in naïve macrophages and with an inhibition of glycolysis in pro-inflammatory macrophages. The aforementioned effects resulted in an increase in M2-like macrophages, which could be reverted by the addition of L-arginine. RNA-seq analysis confirmed alterations in arginase-associated pathways associated with miR-21 antagonism. In conclusion, miR-21 is overexpressed in murine and human CAV, and its targeting delays CAV onset by reprogramming macrophages metabolism.
Collapse
Affiliation(s)
- Vera Usuelli
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Moufida Ben Nasr
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly,Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Francesca D'Addio
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Kaifeng Liu
- Division of Pulmonary and Respiratory DiseasesBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Andrea Vergani
- Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Basset El Essawy
- Department of MedicineAl‐Azhar UniversityCairoEgypt,Renal DivisionTransplantation Research CenterBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Jun Yang
- Institute of Organ TransplantationTongji Hospital and Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Emma Assi
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Mayuko Uehara
- Renal DivisionTransplantation Research CenterBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Chiara Rossi
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Anna Solini
- Department of SurgicalMedical, Molecular and Critical Area PathologyUniversity of PisaPisaItaly
| | - Annalisa Capobianco
- Division of Immunology, Transplantation and Infectious DiseaseSan Raffaele Scientific InstituteMilanItaly
| | - Elena Rigamonti
- Division of Immunology, Transplantation and Infectious DiseaseSan Raffaele Scientific InstituteMilanItaly
| | - Luciano Potena
- Heart Failure and Heart Transplant ProgramS. Orsola‐Malpighi HospitalAlma‐Mater University of BolognaBolognaItaly
| | | | - Mario Sabatino
- Department of Cardiothoracic, Transplantation and Vascular SurgeryS. Orsola‐Malpighi HospitalAlma Mater‐University of BolognaBolognaItaly
| | | | - Enrico Ammirati
- De Gasperis Cardio Center and Transplant CenterNiguarda HospitalMilanItaly
| | - Maria Frigerio
- De Gasperis Cardio Center and Transplant CenterNiguarda HospitalMilanItaly
| | - Eduardo Castillo‐Leon
- Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Anna Maestroni
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Cinzia Azzoni
- Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Cristian Loretelli
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Andy Joe Seelam
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Albert K. Tai
- Tufts University Core Facility (TUCF) Genomics CoreTufts University School of MedicineBostonMassachusetts
| | - Ida Pastore
- Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
| | | | | | - Gary A. Visner
- Division of Pulmonary and Respiratory DiseasesBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Gian V. Zuccotti
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly,Department of PediatricsBuzzi Children's HospitalMilanItaly
| | | | - Reza Abdi
- Renal DivisionTransplantation Research CenterBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Marcus G. Pezzolesi
- Division of Nephrology and Hypertension, Diabetes and Metabolism CenterUniversity of UtahSalt Lake CityUtah
| | - Paolo Fiorina
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly,Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts,Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
| |
Collapse
|
5
|
Glinton K, DeBerge M, Fisher E, Schroth S, Sinha A, Wang JJ, Wasserstrom JA, Ansari MJ, Zhang ZJ, Feinstein M, Leventhal JR, Forbess JM, Lomasney J, Luo X, Thorp EB. Bone marrow-derived AXL tyrosine kinase promotes mitogenic crosstalk and cardiac allograft vasculopathy. J Heart Lung Transplant 2021; 40:435-446. [PMID: 33846079 PMCID: PMC8169599 DOI: 10.1016/j.healun.2021.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 11/19/2022] Open
Abstract
Cardiac Allograft Vasculopathy (CAV) is a leading contributor to late transplant rejection. Although implicated, the mechanisms by which bone marrow-derived cells promote CAV remain unclear. Emerging evidence implicates the cell surface receptor tyrosine kinase AXL to be elevated in rejecting human allografts. AXL protein is found on multiple cell types, including bone marrow-derived myeloid cells. The causal role of AXL from this compartment and during transplant is largely unknown. This is important because AXL is a key regulator of myeloid inflammation. Utilizing experimental chimeras deficient in the bone marrow-derived Axl gene, we report that Axl antagonizes cardiac allograft survival and promotes CAV. Flow cytometric and histologic analyses of Axl-deficient transplant recipients revealed reductions in both allograft immune cell accumulation and vascular intimal thickness. Co-culture experiments designed to identify cell-intrinsic functions of Axl uncovered complementary cell-proliferative pathways by which Axl promotes CAV-associated inflammation. Specifically, Axl-deficient myeloid cells were less efficient at increasing the replication of both antigen-specific T cells and vascular smooth muscle cells (VSMCs), the latter a key hallmark of CAV. For the latter, we discovered that Axl-was required to amass the VSMC mitogen Platelet-Derived Growth Factor. Taken together, our studies reveal a new role for myeloid Axl in the progression of CAV and mitogenic crosstalk. Inhibition of AXL-protein, in combination with current standards of care, is a candidate strategy to prolong cardiac allograft survival.
Collapse
MESH Headings
- Adult
- Animals
- Bone Marrow Cells/metabolism
- Bone Marrow Cells/pathology
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Echocardiography
- Flow Cytometry
- Gene Expression Regulation
- Graft Rejection/diagnosis
- Graft Rejection/genetics
- Graft Rejection/metabolism
- Graft Survival
- Heart Transplantation/adverse effects
- Humans
- Male
- Mice
- Mice, Inbred BALB C
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocytes, Smooth Muscle
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins/genetics
- RNA/genetics
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptor Protein-Tyrosine Kinases/genetics
- Transplantation, Homologous
- Axl Receptor Tyrosine Kinase
Collapse
Affiliation(s)
- Kristofor Glinton
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Matthew DeBerge
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Emily Fisher
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Arjun Sinha
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jiao-Jing Wang
- The Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - J Andrew Wasserstrom
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Mohammed Javeed Ansari
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Zheng Jenny Zhang
- The Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Matthew Feinstein
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Joseph R Leventhal
- The Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Jon Lomasney
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xunrong Luo
- The Department of Nephrology, Duke University School of Medicine, Durham, North Carolina
| | - Edward B Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; The Heart Center, Stanley Manne Children's Research Institute, Lurie Children's Hospital, Chicago, Illinois.
| |
Collapse
|
6
|
Qureshi MS, Alsughayyir J, Chhabra M, Ali JM, Goddard MJ, Devine CA, Conlon TM, Linterman MA, Motallebzadeh R, Pettigrew GJ. Germinal center humoral autoimmunity independently mediates progression of allograft vasculopathy. J Autoimmun 2019; 98:44-58. [DOI: 10.1016/j.jaut.2018.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/18/2022]
|
7
|
Abstract
PURPOSE OF REVIEW Hyperlipidemia is a comorbidity affecting a significant number of transplant patients despite treatment with cholesterol lowering drugs. Recently, it has been shown that hyperlipidemia can significantly alter T-cell responses to cardiac allografts in mice, and graft rejection is accelerated in dyslipidemic mice. Here, we review recent advances in our understanding of hyperlipidemia in graft rejection. RECENT FINDINGS Hyperlipidemic mice have significant increases in serum levels of proinflammatory cytokines, and neutralization of interleukin 17 (IL-17) slows graft rejection, suggesting that IL-17 production by Th17 cells was necessary but not sufficient for rejection. Hyperlipidemia also causes an increase in alloreactive T-cell responses prior to antigen exposure. Analysis of peripheral tolerance mechanisms indicated that this was at least in part due to alterations in FoxP3 T cells that led to reduced Treg function and the expansion of FoxP3 CD4 T cells expressing low levels of CD25. Functionally, alterations in Treg function prevented the ability to induce operational tolerance to fully allogeneic heart transplants through costimulatory-molecule blockade, a strategy that requires Tregs. SUMMARY These findings highlight the importance of considering the contribution of inflammatory comorbidities to cardiac allograft rejection, and point to the potential importance of managing hyperlipidemia in the transplant population.
Collapse
|
8
|
Evolving Approaches in the Identification of Allograft-Reactive T and B Cells in Mice and Humans. Transplantation 2017; 101:2671-2681. [PMID: 28604446 DOI: 10.1097/tp.0000000000001847] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Whether a transplanted allograft is stably accepted, rejected, or achieves immunological tolerance is dependent on the frequency and function of alloreactive lymphocytes, making the identification and analysis of alloreactive T and B cells in transplant recipients critical for understanding mechanisms, and the prediction of allograft outcome. In animal models, tracking the fate of graft-reactive T and B cells allows investigators to uncover their biology and develop new therapeutic strategies to protect the graft. In the clinic, identification and quantification of graft-reactive T and B cells allows for the early diagnosis of immune reactivity and therapeutic intervention to prevent graft loss. In addition to rejection, probing of T and B cell fate in vivo provides insights into the underlying mechanisms of alloimmunity or tolerance that may lead to biomarkers predicting graft fate. In this review, we discuss existing and developing approaches to track and analyze alloreactive T and B cells in mice and humans and provide examples of discoveries made utilizing these techniques. These approaches include mixed lymphocyte reactions, trans-vivo delayed-type hypersensitivity, enzyme-linked immunospot assays, the use of antigen receptor transgenic lymphocytes, and utilization of peptide-major histocompatibility multimers, along with imaging techniques for static multiparameter analysis or dynamic in vivo tracking. Such approaches have already refined our understanding of the alloimmune response and are pointing to new ways to improve allograft outcomes in the clinic.
Collapse
|
9
|
Necroptosis Is Involved in CD4+ T Cell-Mediated Microvascular Endothelial Cell Death and Chronic Cardiac Allograft Rejection. Transplantation 2017; 101:2026-2037. [DOI: 10.1097/tp.0000000000001578] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Ghosh A, Smith M, James SE, Davila ML, Velardi E, Argyropoulos KV, Gunset G, Perna F, Kreines FM, Levy ER, Lieberman S, Jay H, Tuckett AZ, Zakrzewski JL, Tan L, Young LF, Takvorian K, Dudakov JA, Jenq RR, Hanash AM, Motta ACF, Murphy GF, Liu C, Schietinger A, Sadelain M, van den Brink MR. Donor CD19 CAR T cells exert potent graft-versus-lymphoma activity with diminished graft-versus-host activity. Nat Med 2017; 23:242-249. [PMID: 28067900 PMCID: PMC5528161 DOI: 10.1038/nm.4258] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 11/07/2016] [Indexed: 01/09/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative therapy for hematological malignancies. However, graft-versus-host disease (GVHD) and relapse after allo-HSCT remain major impediments to the success of allo-HSCT. Chimeric antigen receptors (CARs) direct tumor cell recognition of adoptively transferred T cells. CD19 is an attractive CAR target, which is expressed in most B cell malignancies, as well as in healthy B cells. Clinical trials using autologous CD19-targeted T cells have shown remarkable promise in various B cell malignancies. However, the use of allogeneic CAR T cells poses a concern in that it may increase risk of the occurrence of GVHD, although this has not been reported in selected patients infused with donor-derived CD19 CAR T cells after allo-HSCT. To understand the mechanism whereby allogeneic CD19 CAR T cells may mediate anti-lymphoma activity without causing a significant increase in the incidence of GVHD, we studied donor-derived CD19 CAR T cells in allo-HSCT and lymphoma models in mice. We demonstrate that alloreactive T cells expressing CD28-costimulated CD19 CARs experience enhanced stimulation, resulting in the progressive loss of both their effector function and proliferative potential, clonal deletion, and significantly decreased occurrence of GVHD. Concurrently, the other CAR T cells that were present in bulk donor T cell populations retained their anti-lymphoma activity in accordance with the requirement that both the T cell receptor (TCR) and CAR be engaged to accelerate T cell exhaustion. In contrast, first-generation and 4-1BB-costimulated CAR T cells increased the occurrence of GVHD. These findings could explain the reduced risk of GVHD occurring with cumulative TCR and CAR signaling.
Collapse
Affiliation(s)
- Arnab Ghosh
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Melody Smith
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Scott E. James
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY,Center of Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marco L. Davila
- Center of Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Enrico Velardi
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kimon V. Argyropoulos
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gertrude Gunset
- Center of Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Fabiana Perna
- Center of Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Fabiana M. Kreines
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Emily R. Levy
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sophie Lieberman
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hilary Jay
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrea Z. Tuckett
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Lisa Tan
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lauren F. Young
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kate Takvorian
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jarrod A. Dudakov
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert R. Jenq
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alan M. Hanash
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ana Carolina F. Motta
- Program in Dermatopathology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - George F. Murphy
- Program in Dermatopathology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, NJ
| | - Andrea Schietinger
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michel Sadelain
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY,Center of Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcel R.M. van den Brink
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY,Center of Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
11
|
Tao YF, Lin F, Yan XY, Gao XG, Teng F, Fu ZR, Wang ZX. Galectin-9 in Combination With EX-527 Prolongs the Survival of Cardiac Allografts in Mice After Cardiac Transplantation. Transplant Proc 2016; 47:2003-9. [PMID: 26293089 DOI: 10.1016/j.transproceed.2015.04.091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/28/2015] [Indexed: 12/23/2022]
Abstract
Galectin-9 (Gal-9), a member of the galectin family, has a variety of biologic activities. However, its role in allografts is not fully clarified yet. The relationship between interleukin-17 (IL-17) and Gal-9 and the role of Gal-9 in T(H)17-cell differentiation also remain unclear. We built a murine cardiac transplantation model, which we treated with Gal-9 and/or EX-527, a specific Sirtuin-1 inhibitor. Afterwards, flow-cytometric analysis and reverse-transcription polymerase chain reaction were used to detect the number of T(H)17 cells and the expression of key factors involved in the differentiation of T(H)17 cells; in addition, the survival times of cardiac transplanted mice in different groups were recorded. The levels of circulating T(H)17 cells were found to increase in the peripheral blood of mice that exhibited acute rejection (AR) after heart transplantation, which was determined to be correlated with the rejection grade. Gal-9 or EX-527 can inhibit the activation and differentiation of T(H)17 cells and effectively suppress T(H)17-cell-mediated AR. These data provide new evidence for the potential regulatory effects of Gal-9 in alloimmune responses in a murine model of heart transplantation, and suggest the combined use of galectin-9 and EX-527 may be a powerful method to induce tolerance of fully mismatched murine cardiac allografts.
Collapse
Affiliation(s)
- Y-f Tao
- Department of General Surgery and Liver Transplant Center, Huashan Hospital, Fudan University, Shanghai, China
| | - F Lin
- Department of General Surgery, Taizhou First People's Hospital, Taizhou, Zhejiang, China
| | - X-y Yan
- Peking University Clinical Research Institute, Health Science Center, Peking University, Beijing, China
| | - X-g Gao
- Division of Liver Transplantation, Organ Transplant Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - F Teng
- Division of Liver Transplantation, Organ Transplant Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Z-r Fu
- Division of Liver Transplantation, Organ Transplant Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Z-x Wang
- Department of General Surgery and Liver Transplant Center, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Yuan J, Bagley J, Iacomini J. Hyperlipidemia Promotes Anti-Donor Th17 Responses That Accelerate Allograft Rejection. Am J Transplant 2015; 15:2336-45. [PMID: 26079335 PMCID: PMC5125017 DOI: 10.1111/ajt.13350] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/13/2015] [Accepted: 02/19/2015] [Indexed: 01/25/2023]
Abstract
Hyperlipidemia occurs in 95% of organ transplant recipients, however its effect on organ allograft rejection has not been investigated. We found that induction of hyperlipidemia in mice caused a significant acceleration of rejection of cardiac allografts. Accelerated rejection was associated with an aggressive T cell infiltrate that mediated significant tissue damage as well as increased serum levels of the proinflammatory cytokines IL-2, IL-6, and IL-17. Hyperlipidemic mice had an increased number of Th17 cells in their periphery and rejecting allografts from hyperlipidemic mice contained significant numbers of IL-17 producing T cells that were not detectable in transplants harvested from controls. Neutralization or genetic ablation of IL-17 prolonged survival of cardiac allografts transplanted into hyperlipidemic recipients, suggesting that IL-17 production promotes accelerated rejection. Analysis of alloreactive T cell frequencies directly ex vivo in naïve mice revealed that the frequency of donor reactive IL-17 producing cells in hyperlipidemic was increased prior to antigen exposure, suggesting that hyperlipidemia was sufficient to alter T cell alloreactivity and promote anti-donor Th17 responses on first exposure to antigen. Together, our data suggest that hyperlipidemia alters rejection by altering the types of T cell subsets that respond to donor antigen by promoting Th17 biased anti-donor reactivity.
Collapse
|
13
|
Bagley J, Yuan J, Chandrakar A, Iacomini J. Hyperlipidemia Alters Regulatory T Cell Function and Promotes Resistance to Tolerance Induction Through Costimulatory Molecule Blockade. Am J Transplant 2015; 15:2324-35. [PMID: 26079467 PMCID: PMC5125018 DOI: 10.1111/ajt.13351] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/13/2015] [Accepted: 03/31/2015] [Indexed: 01/25/2023]
Abstract
Recent work from our laboratory has shown that hyperlipidemia promotes accelerated rejection of vascularized cardiac allografts in mice by inducing anti-donor Th17 reactivity and production of IL-17. Here, we show that hyperlipidemia also affects FoxP3(+) regulatory T cells (Tregs). Hyperlipidemia promotes the development of Tregs that express low levels of CD25. Hyperlipidemia also promotes a decrease in central Tregs and an increase in effector Tregs that appears to account for the increase in the frequency of CD25(low) Tregs. Alterations in Treg subsets also appear to lead to alterations in Treg function. The ability of FoxP3(+) , CD25(high) , CD4(+) Tregs from hyperlipidemic mice to inhibit proliferation of effector T cells stimulated with anti-CD3 and CD28 was reduced when compared with Tregs from control mice. Regulatory T cells isolated from hyperlipidemic recipients exhibit increased activation of Akt, and a reduction in Bim levels that permits the expansion of FoxP3(+) CD25(low) CD4(+) T cells. Hyperlipidemic mice were also resistant to tolerance induction using costimulatory molecule blockade consisting of anti-CD154 and CTLA4Ig, a strategy that requires Tregs. Together, our data suggest that hyperlipidemia profoundly affects Treg subsets and function as well as the ability to induce tolerance.
Collapse
Affiliation(s)
- J. Bagley
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Biomedical Sciences Programs in Immunology and Genetics, Tufts University School of Medicine, Boston, MA
| | - J. Yuan
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Biomedical Sciences Programs in Immunology and Genetics, Tufts University School of Medicine, Boston, MA
| | - A. Chandrakar
- Schuster Family Transplantation Research Center Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - J. Iacomini
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Biomedical Sciences Programs in Immunology and Genetics, Tufts University School of Medicine, Boston, MA,Corresponding author: John Iacomini,
| |
Collapse
|
14
|
Safa K, Ohori S, Borges TJ, Uehara M, Batal I, Shimizu T, Magee CN, Belizaire R, Abdi R, Wu C, Chandraker A, Riella LV. Salt Accelerates Allograft Rejection through Serum- and Glucocorticoid-Regulated Kinase-1-Dependent Inhibition of Regulatory T Cells. J Am Soc Nephrol 2015; 26:2341-7. [PMID: 25833841 DOI: 10.1681/asn.2014090914] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 02/21/2015] [Indexed: 11/03/2022] Open
Abstract
A high-salt diet (HSD) in humans is linked to a number of complications, including hypertension and cardiovascular events. Whether a HSD affects the immune response in transplantation is unknown. Using a murine transplantation model, we investigated the effect of NaCl on the alloimmune response in vitro and in vivo. Incremental NaCl concentrations in vitro augmented T cell proliferation in the settings of both polyclonal and allospecific stimulation. Feeding a HSD to C57BL/6 wild-type recipients of bm12 allografts led to accelerated cardiac allograft rejection, despite similar mean BP and serum sodium levels in HSD and normal salt diet (NSD) groups. The accelerated rejection was associated with a reduction in the proportion of CD4(+)Foxp3(+) regulatory T cells (Tregs) and a significant decrease in Treg proliferation, leading to an increased ratio of antigen-experienced CD4(+) T cells to Tregs in mice recipients of a HSD compared with mice recipients of a NSD. Because serum- and glucocorticoid-regulated kinase-1 (SGK1) has been proposed as a potential target of salt in immune cells, we fed a HSD to CD4(Cre)SGK1(fl/fl) B6-transplanted recipients and observed abrogation of the deleterious effect of a HSD in the absence of SGK1 on CD4(+) cells. In summary, we show that NaCl negatively affects the regulatory balance of T cells in transplantation and precipitates rejection in an SGK1-dependent manner.
Collapse
Affiliation(s)
- Kassem Safa
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Transplant Center and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shunsuke Ohori
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thiago J Borges
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; School of Biosciences and Biomedical Research Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Mayuko Uehara
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ibrahim Batal
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Tetsunosuke Shimizu
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ciara N Magee
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Renal Medicine and Transplantation, Royal Free London, National Health Service Foundation Trust, London, United Kingdom; and
| | - Roger Belizaire
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Reza Abdi
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chuan Wu
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anil Chandraker
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Leonardo V Riella
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
15
|
Yeung MY, Ding Q, Brooks CR, Xiao S, Workman CJ, Vignali DA, Ueno T, Padera RF, Kuchroo VK, Najafian N, Rothstein DM. TIM-1 signaling is required for maintenance and induction of regulatory B cells. Am J Transplant 2015; 15:942-53. [PMID: 25645598 PMCID: PMC4530122 DOI: 10.1111/ajt.13087] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/09/2014] [Accepted: 10/28/2014] [Indexed: 01/25/2023]
Abstract
Apart from their role in humoral immunity, B cells can exhibit IL-10-dependent regulatory activity (Bregs). These regulatory subpopulations have been shown to inhibit inflammation and allograft rejection. However, our understanding of Bregs has been hampered by their rarity, lack of a specific marker, and poor insight into their induction and maintenance. We previously demonstrated that T cell immunoglobulin mucin domain-1 (TIM-1) identifies over 70% of IL-10-producing B cells, irrespective of other markers. We now show that TIM-1 is the primary receptor responsible for Breg induction by apoptotic cells (ACs). However, B cells that express a mutant form of TIM-1 lacking the mucin domain (TIM-1(Δmucin) ) exhibit decreased phosphatidylserine binding and are unable to produce IL-10 in response to ACs or by specific ligation with anti-TIM-1. TIM-1(Δmucin) mice also exhibit accelerated allograft rejection, which appears to be due in part to their defect in both baseline and induced IL-10(+) Bregs, since a single transfer of WT TIM-1(+) B cells can restore long-term graft survival. These data suggest that TIM-1 signaling plays a direct role in Breg maintenance and induction both under physiological conditions (in response to ACs) and in response to therapy through TIM-1 ligation. Moreover, they directly demonstrate that the mucin domain regulates TIM-1 signaling.
Collapse
Affiliation(s)
- Melissa Y. Yeung
- Transplantation Research Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Qing Ding
- Thomas E. Starzl Transplantation Institute, Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Craig R. Brooks
- Renal Division, Harvard Medical School, Boston, Massachusetts, USA
| | - Sheng Xiao
- Center for Neurologic Disease, Harvard Medical School, Boston, Massachusetts, USA
| | - Creg J. Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Dario A.A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Takuya Ueno
- Transplantation Research Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert F. Padera
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vijay K. Kuchroo
- Center for Neurologic Disease, Harvard Medical School, Boston, Massachusetts, USA
| | - Nader Najafian
- Transplantation Research Center, Harvard Medical School, Boston, Massachusetts, USA
- Department of Nephrology, Cleveland Clinic Florida, Weston, FL, USA
| | - David M. Rothstein
- Thomas E. Starzl Transplantation Institute, Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Tregitope: Immunomodulation powerhouse. Hum Immunol 2014; 75:1139-46. [PMID: 25454619 DOI: 10.1016/j.humimm.2014.10.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 07/07/2014] [Accepted: 10/09/2014] [Indexed: 01/29/2023]
Abstract
IVIG is frequently used in the 'pre-conditioning' regimens for higher risk transplants; its effects are attributed in part to induction of Tregs. We have identified regulatory T cell (Treg) epitopes, now known as Tregitopes, in IgG, the main component of intravenous immunoglobulin therapy (IVIg). Tregitopes provide one explanation for the expansion and activation of Treg cells following IVIg treatment. Tregitopes are peptides that exhibit high affinity binding to multiple human HLA Class II DR; they are conserved across IgG isotypes and mammalian species. In vitro and in vivo, for human PBMC and in animal models, Tregitopes activate Tregs. Studies to delineate the mechanism of action have shown that Tregitopes' effects are very similar to IVIg in vitro. Here we demonstrate that Tregitopes induce Tregs to produce IL-10, leading to modulation of dendritic cell phenotype (down-regulation of Class II, CD80 and CD86 and up-regulation of ILT3), and describe the effects of Tregitopes in the ABM-TCR-transgenic skin transplantation model. The discovery of Tregitopes in IgG and other autologous proteins may contribute to improved understanding of the mechanism of action of IVIg and lead to the application of these powerful immunomodulators to improve transplantation success and suppress autoimmune disease, in the future.
Collapse
|
17
|
Zou H, Yang Y, Gao M, Zhang B, Ming B, Sun Y, Chen H, Tang X, Chen Z, Xiong P, Xu Y, Fang M, Tan Z, Gong F, Zheng F. HMGB1 is involved in chronic rejection of cardiac allograft via promoting inflammatory-like mDCs. Am J Transplant 2014; 14:1765-77. [PMID: 24984831 DOI: 10.1111/ajt.12781] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 04/04/2014] [Accepted: 04/06/2014] [Indexed: 01/25/2023]
Abstract
Chronic rejection that leads to diffuse narrowing and occlusion of graft vessels is the most important cause of morbidity and mortality following cardiac transplantation. The role and underlying mechanism of high-mobility group box 1 (HMGB1), as an established inflammatory mediator in acute rejection, remains poorly understood in chronic rejection. Here, we assessed the effects and mechanisms of HMGB1 on the chronic rejection using single MHC Class II-mismatched mouse cardiac transplantation model. It was found that HMGB1 was increased accompanying with the development of chronic rejection, while blockade of HMGB1 with specific neutralizing mAb substantially ameliorated chronic rejection-mediated vasculopathy and fibrosis of allograft, as well as markedly decreased T cell infiltration and production of IL-17A and interferon-gamma in allograft and recipient's spleen. Further, anti-HMGB1 antibody treatment significantly declined the number and frequency of mature dendritic cells (DCs) in allograft and recipient's spleen, especially CD11b(+) Ly6C(high) matured DCs that share the phenotypes with inflammatory-DCs. These findings indicate that HMGB1 contributes to chronic rejection, and HMGB1 blockade may be a novel mean to disrupt the proinflammatory loop after heart transplantation.
Collapse
Affiliation(s)
- H Zou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, China; Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wang S, Xu X, Xie A, Li J, Ye P, Liu Z, Wu J, Rui L, Xia J. Anti-interleukin-12/23p40 antibody attenuates chronic rejection of cardiac allografts partly via inhibition γδT cells. Clin Exp Immunol 2012; 169:320-9. [PMID: 22861372 DOI: 10.1111/j.1365-2249.2012.04612.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In our previous study, we showed that treatment with an anti-interleukin (IL)-12/23p40 antibody inhibits acute cardiac allograft rejection via inhibiting production of interferon (IFN)-γ and IL-17a. However, the impact of this antagonistic anti-p40 antibody on chronic cardiac rejection was unclear. Hearts of B6.C-H2bm12/KhEg mice were transplanted into major histocompatibility complex (MHC) class II-mismatched C57Bl/6J mice (wild-type, γδTCR (-/-) and IL-17(-/-) ), which is an established murine model of chronic allograft rejection without immunosuppression. The mice were treated with control immunoglobulin (Ig)G or 200 µg anti-p40 monoclonal antibody on post-operative days, respectively. Abdominal palpation and echocardiography were used to monitor graft survival. The mice administered with anti-p40 antibody showed a significant promotion in graft survival (median survival time >100 days), and histological analyses revealed that cardiac allograft rejection was attenuated. Quantitative real-time polymerase chain reaction (qRT-PCR) and immunofluorescence analyses demonstrated that anti-p40 antibody down-regulated the level of ingraft cytokine and chemokine expression (IL-6, IFN-γ, IL-17a, CCL2 and CCL20). Flow cytometry analyses showed that γδ T cells are an important ingraft source of IFN-γ and IL-17a and inhibit the production of inflammation cytokine by anti-p40 antibody. Compared with the wild-type group, the graft survival time in the γδ T cell receptor(-/-) and IL-17(-/-) mice was prolonged significantly. Therefore we propose that, in the chronic allograft rejection model, treatment with anti-p40 antibody prolongs graft survival possibly by reducing the amount of reactive inflammatory cells, especially γδ T cells.
Collapse
Affiliation(s)
- S Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Institute of Cardiovascular Surgery, Xijing Hospital, The 4th Military Medical University, Xi'an, Shaanxi Province Department of Cardiovascular Surgery, Fu Wai Hospital, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Jenq RR, Ubeda C, Taur Y, Menezes CC, Khanin R, Dudakov JA, Liu C, West ML, Singer NV, Equinda MJ, Gobourne A, Lipuma L, Young LF, Smith OM, Ghosh A, Hanash AM, Goldberg JD, Aoyama K, Blazar BR, Pamer EG, van den Brink MRM. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. ACTA ACUST UNITED AC 2012; 209:903-11. [PMID: 22547653 PMCID: PMC3348096 DOI: 10.1084/jem.20112408] [Citation(s) in RCA: 484] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
GVHD is associated with significant shifts in the composition of the intestinal microbiota in human and mouse models; manipulating the microbiota can alter the severity of GVHD in mice. Despite a growing understanding of the link between intestinal inflammation and resident gut microbes, longitudinal studies of human flora before initial onset of intestinal inflammation have not been reported. Here, we demonstrate in murine and human recipients of allogeneic bone marrow transplantation (BMT) that intestinal inflammation secondary to graft-versus-host disease (GVHD) is associated with major shifts in the composition of the intestinal microbiota. The microbiota, in turn, can modulate the severity of intestinal inflammation. In mouse models of GVHD, we observed loss of overall diversity and expansion of Lactobacillales and loss of Clostridiales. Eliminating Lactobacillales from the flora of mice before BMT aggravated GVHD, whereas reintroducing the predominant species of Lactobacillus mediated significant protection against GVHD. We then characterized gut flora of patients during onset of intestinal inflammation caused by GVHD and found patterns mirroring those in mice. We also identified increased microbial chaos early after allogeneic BMT as a potential risk factor for subsequent GVHD. Together, these data demonstrate regulation of flora by intestinal inflammation and suggest that flora manipulation may reduce intestinal inflammation and improve outcomes for allogeneic BMT recipients.
Collapse
Affiliation(s)
- Robert R Jenq
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Carvello M, Petrelli A, Vergani A, Lee KM, Tezza S, Chin M, Orsenigo E, Staudacher C, Secchi A, Dunussi-Joannopoulos K, Sayegh MH, Markmann JF, Fiorina P. Inotuzumab ozogamicin murine analog-mediated B-cell depletion reduces anti-islet allo- and autoimmune responses. Diabetes 2012; 61:155-65. [PMID: 22076927 PMCID: PMC3237644 DOI: 10.2337/db11-0684] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
B cells participate in the priming of the allo- and autoimmune responses, and their depletion can thus be advantageous for islet transplantation. Herein, we provide an extensive study of the effect of B-cell depletion in murine models of islet transplantation. Islet transplantation was performed in hyperglycemic B-cell-deficient(μMT) mice, in a purely alloimmune setting (BALB/c into hyperglycemic C57BL/6), in a purely autoimmune setting (NOD.SCID into hyperglycemic NOD), and in a mixed allo-/autoimmune setting (BALB/c into hyperglycemic NOD). Inotuzumab ozogamicin murine analog (anti-CD22 monoclonal antibody conjugated with calicheamicin [anti-CD22/cal]) efficiently depleted B cells in all three models of islet transplantation examined. Islet graft survival was significantly prolonged in B-cell-depleted mice compared with control groups in transplants of islets from BALB/c into C57BL/6 (mean survival time [MST]: 16.5 vs. 12.0 days; P = 0.004), from NOD.SCID into NOD (MST: 23.5 vs. 14.0 days; P = 0.03), and from BALB/c into NOD (MST: 12.0 vs. 5.5 days; P = 0.003). In the BALB/c into B-cell-deficient mice model, islet survival was prolonged as well (MST: μMT = 32.5 vs. WT = 14 days; P = 0.002). Pathology revealed reduced CD3(+) cell islet infiltration and confirmed the absence of B cells in treated mice. Mechanistically, effector T cells were reduced in number, concomitant with a peripheral Th2 profile skewing and ex vivo recipient hyporesponsiveness toward donor-derived antigen as well as islet autoantigens. Finally, an anti-CD22/cal and CTLA4-Ig-based combination therapy displayed remarkable prolongation of graft survival in the stringent model of islet transplantation (BALB/c into NOD). Anti-CD22/cal-mediated B-cell depletion promotes the reduction of the anti-islet immune response in various models of islet transplantation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antibody-Dependent Cell Cytotoxicity/physiology
- Autoimmunity/drug effects
- B-Lymphocytes/cytology
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- Cell Death/drug effects
- Cell Death/immunology
- Cells, Cultured
- Female
- Graft Survival/immunology
- Inotuzumab Ozogamicin
- Islets of Langerhans/drug effects
- Islets of Langerhans/immunology
- Islets of Langerhans Transplantation/immunology
- Islets of Langerhans Transplantation/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Transplantation Tolerance/drug effects
- Transplantation Tolerance/immunology
Collapse
Affiliation(s)
- Michele Carvello
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Petrelli
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Vergani
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kang Mi Lee
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sara Tezza
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Melissa Chin
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Orsenigo
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Carlo Staudacher
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Secchi
- Department of Medicine, San Raffaele Scientific Institute, Milan, Italy
| | | | - Mohamed H. Sayegh
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - James F. Markmann
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paolo Fiorina
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, San Raffaele Scientific Institute, Milan, Italy
- Corresponding author: Paolo Fiorina,
| |
Collapse
|
21
|
D'Addio F, Riella LV, Mfarrej BG, Chabtini L, Adams LT, Yeung M, Yagita H, Azuma M, Sayegh MH, Guleria I. The link between the PDL1 costimulatory pathway and Th17 in fetomaternal tolerance. THE JOURNAL OF IMMUNOLOGY 2011; 187:4530-41. [PMID: 21949023 DOI: 10.4049/jimmunol.1002031] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Fetomaternal tolerance has been shown to depend both on regulatory T cells (Tregs) and negative signals from the PD1-PDL1 costimulatory pathway. More recently, IL-17-producing T cells (Th17) have been recognized as a barrier in inducing tolerance in transplantation. In this study, we investigate the mechanisms of PDL1-mediated regulation of fetomaternal tolerance using an alloantigen-specific CD4(+) TCR transgenic mouse model system (ABM-tg mouse). PDL1 blockade led to an increase in embryo resorption and a reduction in litter size. This was associated with a decrease in Tregs, leading to a lower Treg/effector T cell ratio. Moreover, PDL1 blockade inhibited Ag-specific alloreactive T cell apoptosis and induced apoptosis of Tregs and a shift toward higher frequency of Th17 cells, breaking fetomaternal tolerance. These Th17 cells arose predominantly from CD4(+)Foxp3(-) cells, rather than from conversion of Tregs. Locally in the placenta, similar decrease in regulatory and apoptotic markers was observed by real-time PCR. Neutralization of IL-17 abrogated the anti-PDL1 effect on fetal survival rate and restored Treg numbers. Finally, the adoptive transfer of Tregs was also able to improve fetal survival in the setting of PDL1 blockade. This is to our knowledge the first report using an alloantigen-specific model that establishes a link between PDL1, Th17 cells, and fetomaternal tolerance.
Collapse
Affiliation(s)
- Francesca D'Addio
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School Renal Division, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Brunner SM, Schiechl G, Falk W, Schlitt HJ, Geissler EK, Fichtner-Feigl S. Interleukin-33 prolongs allograft survival during chronic cardiac rejection. Transpl Int 2011; 24:1027-39. [PMID: 21797940 DOI: 10.1111/j.1432-2277.2011.01306.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Interleukin-33 (IL-33) stimulates the generation of cells and cytokines characteristic of a Th2 immune response. We examined the effects of IL-33 on allografted heart tissue in a chronic cardiac rejection model, including analysis of the peripheral myeloid and lymphoid compartments. B6.C-H2bm12/KhEg hearts were transplanted into MHC class II-mismatched C57Bl/6J mice; IL-33 was administered daily. Cells from allografts and spleens were isolated for flow cytometry and cultured for cytokine production; some tissues were used for immunohistochemistry. Animals treated with IL-33 showed significantly longer allograft survival, which was associated with a distinct cytokine profile produced by graft-infiltrating cells. Proinflammatory IL-17A production was decreased with IL-33 treatment, while increased levels of IL-5, IL-10, and IL-13 were observed. After IL-33 therapy, flow cytometry showed a direct induction of CD4(+) Foxp3(+) Treg, whereas the number of B220(+) CD19(+) B cells, and circulating, as well as allograft deposited, alloantibodies was reduced. Following IL-33 treatment, a significant decrease in graft-infiltrating CD11b(high) Gr1(high) granulocytes coincided with a significant increase in CD11b(high) Gr1(intermediate) myeloid-derived suppressor cells (MDSC). In conclusion, IL-33 treatment in the setting of chronic rejection promotes the development of a Th2-type immune response that favors MDSC and Treg expansion, reduces antibody-mediated rejection (AMR), and ultimately, prolongs allograft survival.
Collapse
Affiliation(s)
- Stefan M Brunner
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Animal models of chronic allograft injury: contributions and limitations to understanding the mechanism of long-term graft dysfunction. Transplantation 2010; 90:935-44. [PMID: 20703180 DOI: 10.1097/tp.0b013e3181efcfbc] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Advances in immunosuppression have reduced the incidence of acute graft loss after transplantation, but long-term allograft survival is still hindered by the development of chronic allograft injury, a multifactorial process that involves both immunologic and nonimmunologic components. Because these components become defined in the clinical setting, development of animal models enables exploration into underlying mechanisms leading to long-term graft dysfunction. This review presents animal models that have enabled investigation into chronic allograft injury and discusses pivotal models currently being used. The mechanisms uncovered by these models will ultimately lead to development of new therapeutic options to prevent long-term graft dysfunction.
Collapse
|
24
|
A novel clinically relevant approach to tip the balance toward regulation in stringent transplant model. Transplantation 2010; 90:260-9. [PMID: 20712076 DOI: 10.1097/tp.0b013e3181e64217] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Regulatory T cells (Tregs) actively regulate alloimmune responses and promote transplantation tolerance. Thymoglobulin, a rabbit polyclonal antithymocyte globulin (ATG), is a widely used induction therapy in clinical organ transplantation that depletes peripheral T cells. However, resistance to tolerance induction is seen with certain T-cell depleting strategies and is attributed to alterations in the balance of naive, memory and Tregs. The exact mechanism of action of ATG and its effects on the homeostasis and balance between Tregs and T-effector-memory cells (Tem) are unknown. METHODS A novel antibody reagent, rabbit polyclonal anti-murine thymocyte globulin (mATG), generated by the same process used to manufacture thymoglobulin, was used alone or in combination with CTLA4Ig or sirolimus (SRL) in a stringent fully major histocompatibility complex-mismatched murine skin allograft model to study graft survival and mechanisms involved. RESULTS mATG depletes T cells but preferentially spares CD25+ natural Tregs which limit skewing of T-cell repertoire toward Tem phenotype among the recovering T cells. T-cell depletion with mATG combined with CTLA4Ig and SRL synergize to prolong graft survival by tipping the Treg/Tem balance further in favor of Tregs by preserving Tregs, facilitating generation of new Tregs by a conversion mechanism and limiting Tem expansion in response to alloantigen and homeostatic proliferation. CONCLUSIONS Simultaneous T-cell depletion with ATG and costimulatory blockade, combined with SRL, synergizes to promote regulation and prolong allograft survival in a stringent transplant model. These results provide the rationale for translating such novel combination therapy to promote regulation in primate and human organ transplantation.
Collapse
|
25
|
Yamaura K, Boenisch O, Watanabe T, Ueno T, Vanguri V, Yang J, Tanaka K, Guleria I, Borst J, Zhai Y, Kupiec-Weglinski JW, Najafian N. Differential requirement of CD27 costimulatory signaling for naïve versus alloantigen-primed effector/memory CD8+ T cells. Am J Transplant 2010; 10:1210-20. [PMID: 20353477 PMCID: PMC2889922 DOI: 10.1111/j.1600-6143.2010.03089.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CD8(+) memory T cells endanger allograft survival by causing acute and chronic rejection and prevent tolerance induction. We explored the role of CD27:CD70 T-cell costimulatory pathway in alloreactive CD8(+)/CD4(+) T-cell activation. CD27-deficient (CD27(-/-)) and wild-type (WT) B6 mice rejected BALB/c cardiac allografts at similar tempo, with or without depletion of CD4(+) or CD8(+) T cells, suggesting that CD27 is not essential during primary T-cell alloimmune responses. To dissect the role of CD27 in primed effector and memory alloreactive T cells, CD27(-/-) or WT mice were challenged with BALB/c hearts either 10 or 40 days after sensitization with donor-type skin grafts. Compared to WT controls, allograft survival was prolonged in day 40- but not day 10-sensitized CD27(-/-) recipients. Improved allograft survival was accompanied by diminished secondary responsiveness of memory CD8(+) T cells, which resulted from deficiency in memory formation rather than their lack of secondary expansion. Chronic allograft vasculopathy and fibrosis were diminished in CD27(-/-) recipients of class I- but not class II-mismatched hearts as compared to WT controls. These data establish a novel role for CD27 as an important costimulatory molecule for alloreactive CD8(+) memory T cells in acute and chronic allograft rejection.
Collapse
Affiliation(s)
- K. Yamaura
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital, Harvard Medical School, Boston, MA
| | - O. Boenisch
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital, Harvard Medical School, Boston, MA
| | - T. Watanabe
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital, Harvard Medical School, Boston, MA
| | - T. Ueno
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital, Harvard Medical School, Boston, MA
| | - V. Vanguri
- Immunology Research Division, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - J. Yang
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital, Harvard Medical School, Boston, MA
| | - K. Tanaka
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital, Harvard Medical School, Boston, MA
| | - I. Guleria
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital, Harvard Medical School, Boston, MA
| | - J. Borst
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Y. Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - J. W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - N. Najafian
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital, Harvard Medical School, Boston, MA,Corresponding author: Nader Najafian,
| |
Collapse
|
26
|
Win TS, Rehakova S, Negus MC, Saeb-Parsy K, Goddard M, Conlon TM, Bolton EM, Bradley JA, Pettigrew GJ. Donor CD4 T cells contribute to cardiac allograft vasculopathy by providing help for autoantibody production. Circ Heart Fail 2009; 2:361-9. [PMID: 19808360 DOI: 10.1161/circheartfailure.108.827139] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The development of autoantibody after heart transplantation is increasingly associated with poor graft outcome, but what triggers its development and whether it has a direct causative role in graft rejection is not clear. Here, we study the development of antinuclear autoantibody in an established mouse model of heart allograft vasculopathy. METHODS AND RESULTS Humoral vascular changes, including endothelial complement staining, were present in bm12 heart grafts, explanted 50 days after transplantation. Alloantibody was not detectable, but long-lasting autoantibody responses developed in C57BL/6 recipients from the third week after transplantation. No autoantibody was generated if donor CD4 T cells were depleted before heart graft retrieval or in recipients that lacked B-cell major histocompatibility complex class II expression, indicating that humoral autoimmunity is a consequence of donor CD4 T-cell allorecognition of the major histocompatibility complex class II complex on recipient autoreactive B cells. An effector role for autoantibody in graft rejection was confirmed by abrogation of humoral vascular rejection, and attenuation of vasculopathy, in B-cell deficient recipients and by development of vascular obliteration and accelerated rejection in recipients primed for autoantibody before transplantation. CONCLUSIONS Passenger CD4 T cells within heart transplants can contribute to allograft vasculopathy by providing help to recipient B cells for autoantibody generation.
Collapse
Affiliation(s)
- Thet Su Win
- Department of Surgery, Addenbrooke's Hospital, Cambridge, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yuan X, Paez-Cortez J, Schmitt-Knosalla I, D'Addio F, Mfarrej B, Donnarumma M, Habicht A, Clarkson MR, Iacomini J, Glimcher LH, Sayegh MH, Ansari MJ. A novel role of CD4 Th17 cells in mediating cardiac allograft rejection and vasculopathy. ACTA ACUST UNITED AC 2008; 205:3133-44. [PMID: 19047438 PMCID: PMC2605226 DOI: 10.1084/jem.20081937] [Citation(s) in RCA: 239] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
T-bet plays a crucial role in Th1 development. We investigated the role of T-bet in the development of allograft rejection in an established MHC class II–mismatched (bm12 into B6) model of chronic allograft vasculopathy (CAV). Intriguingly, and in contrast to IFN-γ−/− mice that are protected from CAV, T-bet−/− recipients develop markedly accelerated allograft rejection accompanied by early severe vascular inflammation and vasculopathy, and infiltration by predominantly IL-17–producing CD4 T cells. Concurrently, T-bet−/− mice exhibit a T helper type 1 (Th1)–deficient environment characterized by profound IFN-γ deficiency, a Th2 switch characterized by increased production of interleukin (IL) 4, IL-5, IL-10, and IL-13 cytokines, as well as increased production of the proinflammatory cytokines IL-6, IL-12p40, and IL-17. Neutralization of IL-17 inhibits accelerated allograft rejection and vasculopathy in T-bet−/− mice. Interestingly, CD4 but not CD8 T cell deficiency in T-bet−/− mice affords dramatic protection from vasculopathy and facilitates long-term graft acceptance. This is the first study establishing that in the absence of Th1-mediated alloimmune responses, CD4 Th17 cells mediate an aggressive proinflammatory response culminating in severe accelerated allograft rejection and vasculopathy. These results have important implications for the development of novel therapies to target this intractable problem in clinical solid organ transplantation.
Collapse
Affiliation(s)
- Xueli Yuan
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
A new T-cell receptor transgenic model of the CD4+ direct pathway: level of priming determines acute versus chronic rejection. Transplantation 2008; 85:247-55. [PMID: 18212630 DOI: 10.1097/tp.0b013e31815e883e] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND T-cell receptor transgenic (TCR-tg) mouse models with direct CD4 alloreactivity will help elucidate mechanisms of transplant rejection and tolerance in vivo. Although such models exist, they are limited by unusual strain combinations or are based on model antigens. METHODS A TCR-tg mouse with direct CD4 specificity in the widely used BALB/c donor --> C57BL/6 host strain combination was created. This TCR-tg mouse, named 4C, was selected for reactivity against BALB/c dendritic cells in order to model early priming events after transplantation. The response of 4C T cells to skin and heart transplants were characterized. RESULTS The alloantigen is restricted by I-A and appears to be widely distributed in mouse tissues. 4C T cells are able to acutely reject skin but not heart allografts. Paradoxically, heart grafts elicited a stronger proliferation and effector function of TCR-tg T cells than skin grafts. 4C T cells caused cardiac allograft vasculopathy in the absence of other T cells and alloantibodies, suggesting a role for the direct pathway in chronic rejection. Augmentation of priming with an infusion of donor-derived dendritic cells resulted in acute heart allograft rejection by 4C T cells, demonstrating that the level of priming can play a role in determining acute versus chronic rejection by the CD4 direct pathway. CONCLUSIONS Rejection of a graft by the direct CD4 pathway is determined by graft susceptibility to rejection, as well as the degree of T-cell priming caused by the graft. Grafts that are not acutely rejected can develop transplant vasculopathy mediated by the direct CD4 T cells.
Collapse
|
29
|
Ueno T, Habicht A, Clarkson MR, Albin MJ, Yamaura K, Boenisch O, Popoola J, Wang Y, Yagita H, Akiba H, Ansari MJ, Yang J, Turka LA, Rothstein DM, Padera RF, Najafian N, Sayegh MH. The emerging role of T cell Ig mucin 1 in alloimmune responses in an experimental mouse transplant model. J Clin Invest 2008; 118:742-51. [PMID: 18172549 DOI: 10.1172/jci32451] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Accepted: 11/19/2007] [Indexed: 12/20/2022] Open
Abstract
T cell Ig mucin 1 (TIM-1) plays an important role in regulating immune responses in autoimmune and asthma models, and it is expressed on both Th1 and Th2 cells. Using an antagonistic TIM-1-specific antibody, we studied the role of TIM-1 in alloimmunity. A short course of TIM-1-specific antibody monotherapy prolonged survival of fully MHC-mismatched vascularized mouse cardiac allografts. This prolongation was associated with inhibition of alloreactive Th1 responses and preservation of Th2 responses. TIM-1-specific antibody treatment was more effective in Th1-type cytokine-deficient Stat4(-/-) recipients as compared with Th2-type cytokine-deficient Stat6(-/-) recipients. Subtherapeutic doses of rapamycin plus TIM-1-specific antibody resulted in allograft acceptance and prevented the development of chronic allograft vasculopathy. Allograft survival via this treatment was accompanied by a Th1- to Th2-type cytokine switch. Depletion of natural Tregs abrogated the graft-protecting effect of the TIM-1-specific antibody. Importantly, CD4(+)CD25(+) Tregs obtained from long-term survivors had enhanced regulatory activity as compared with naive CD4(+)CD25(+) Tregs. Consistent with this, TIM-1-specific antibody treatment both preserved Tregs and prevented the expansion of alloreactive effector Th1 cells in an alloreactive TCR transgenic adoptive transfer model. These studies define previously unknown functions of TIM-1 in regulating alloimmune responses in vivo and may provide a novel approach to promoting transplantation tolerance.
Collapse
Affiliation(s)
- Takuya Ueno
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yang J, Popoola J, Khandwala S, Vadivel N, Vanguri V, Yuan X, Dada S, Guleria I, Tian C, Ansari MJ, Shin T, Yagita H, Azuma M, Sayegh MH, Chandraker A. Critical role of donor tissue expression of programmed death ligand-1 in regulating cardiac allograft rejection and vasculopathy. Circulation 2008; 117:660-9. [PMID: 18212277 DOI: 10.1161/circulationaha.107.741025] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Allograft vasculopathy is a major limiting factor in the long-term success of cardiac transplantation. T cells play a critical role in initiation of cardiac allograft rejection and allograft vasculopathy. The negative T-cell costimulatory pathway PD-1:PDL1/PDL2 (programmed death-1:programmed death ligand-1/2) plays an important role in regulating alloimmune responses. We investigated the role of recipient versus donor PD-1 ligands in the pathogenesis of allograft rejection with emphasis on the role of tissue expression in regulating this alloimmune response in vivo. METHODS AND RESULTS We used established major histocompatibility complex class II- and class I-mismatched models of vascularized cardiac allograft rejection, blocking anti-PDL1 and anti-PDL2 antibodies, and PDL1- and PDL2-deficient mice (as donors or recipients) to study the role of the PD-1:PDL1/PDL2 pathway in chronic rejection. We also used PDL1-deficient and wild-type mice and bone marrow transplantation to generate chimeric animals that express PDL1 exclusively on either hematopoietic or parenchymal cells. PDL1 but not PDL2 blockade significantly accelerated cardiac allograft rejection in the bm12-into-B6 and B6-into-bm12 models. Although wild-type cardiac allografts survived long term, PDL1-/- donor hearts transplanted into wild-type bm12 mice exhibited accelerated rejection and vasculopathy associated with enhanced recipient T-cell alloreactivity. Interestingly, PDL1-/- recipients did not exhibit an accelerated tempo of cardiac allograft rejection. Using chimeric animals as donors, we show that PDL1 expression on cardiac tissue alone significantly prolonged graft survival compared with full PDL1-/- donor grafts in transplanted wild-type recipients. CONCLUSIONS This is the first report to demonstrate that expression of the negative costimulatory molecule PDL1 on donor cardiac tissue regulates recipient alloimmune responses, allograft rejection, and vasculopathy.
Collapse
Affiliation(s)
- Jun Yang
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Tanaka K, Albin MJ, Yuan X, Yamaura K, Habicht A, Murayama T, Grimm M, Waaga AM, Ueno T, Padera RF, Yagita H, Azuma M, Shin T, Blazar BR, Rothstein DM, Sayegh MH, Najafian N. PDL1 is required for peripheral transplantation tolerance and protection from chronic allograft rejection. THE JOURNAL OF IMMUNOLOGY 2007; 179:5204-10. [PMID: 17911605 PMCID: PMC2291549 DOI: 10.4049/jimmunol.179.8.5204] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The PD-1:PDL pathway plays an important role in regulating alloimmune responses but its role in transplantation tolerance is unknown. We investigated the role of PD-1:PDL costimulatory pathway in peripheral and a well established model of central transplantation tolerance. Early as well as delayed blockade of PDL1 but not PDL2 abrogated tolerance induced by CTLA4Ig in a fully MHC-mismatched cardiac allograft model. Accelerated rejection was associated with a significant increase in the frequency of IFN-gamma-producing alloreactive T cells and expansion of effector CD8(+) T cells in the periphery, and a decline in the percentage of Foxp3(+) graft infiltrating cells. Similarly, studies using PDL1/L2-deficient recipients confirmed the results with Ab blockade. Interestingly, while PDL1-deficient donor allografts were accepted by wild-type recipients treated with CTLA4Ig, the grafts developed severe chronic rejection and vasculopathy when compared with wild-type grafts. Finally, in a model of central tolerance induced by mixed allogeneic chimerism, engraftment was not abrogated by PDL1/L2 blockade. These novel data demonstrate the critical role of PDL1 for induction and maintenance of peripheral transplantation tolerance by its ability to alter the balance between pathogenic and regulatory T cells. Expression of PDL1 in donor tissue is critical for prevention of in situ graft pathology and chronic rejection.
Collapse
Affiliation(s)
- Katsunori Tanaka
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Felix NJ, Donermeyer DL, Horvath S, Walters JJ, Gross ML, Suri A, Allen PM. Alloreactive T cells respond specifically to multiple distinct peptide-MHC complexes. Nat Immunol 2007; 8:388-97. [PMID: 17322886 DOI: 10.1038/ni1446] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Accepted: 01/31/2007] [Indexed: 01/07/2023]
Abstract
The molecular basis underlying the specificity of alloreactive T cells for peptide-major histocompatibility complex ligands has been elusive. Here we describe a screen of 60 I-E(k)-alloreactive T cells and 83 naturally processed peptides that identified 9 reactive T cells. Three of the T cells responded to multiple, distinct peptides that shared no sequence homology. These T cells recognized each peptide-major histocompatibility complex ligand specifically and used a distinct constellation of I-E(k) contact residues for each interaction. Our studies show that alloreactive T cells have a 'germline-encoded' capacity to recognize multiple, distinct ligands and thus show 'polyspecificity', not degeneracy. Our findings help to explain the high frequency of alloreactive T cells and provide insight into the nature of T cell specificity.
Collapse
Affiliation(s)
- Nathan J Felix
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63130, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Ford ML, Koehn BH, Wagener ME, Jiang W, Gangappa S, Pearson TC, Larsen CP. Antigen-specific precursor frequency impacts T cell proliferation, differentiation, and requirement for costimulation. ACTA ACUST UNITED AC 2007; 204:299-309. [PMID: 17261633 PMCID: PMC2118720 DOI: 10.1084/jem.20062319] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
After a brief period of antigenic stimulation, T cells become committed to a program of autonomous expansion and differentiation. We investigated the role of antigen-specific T cell precursor frequency as a possible cell-extrinsic factor impacting T cell programming in a model of allogeneic tissue transplantation. Using an adoptive transfer system to incrementally raise the precursor frequency of antigen-specific CD8+ T cells, we found that donor-reactive T cells primed at low frequency exhibited increased cellular division, decreased development of multifunctional effector activity, and an increased requirement for CD28- and CD154-mediated costimulation relative to those primed at high frequency. The results demonstrated that recipients with low CD4+ and CD8+ donor-reactive T cell frequencies exhibited long-term skin graft survival upon CD28/CD154 blockade, whereas simultaneously raising the frequency of CD4+ T cells to ∼0.5% and CD8+ T cells to ∼5% precipitated graft rejection despite CD28/CD154 blockade. Antigenic rechallenge of equal numbers of cells stimulated at high or low frequency revealed that cells retained an imprint of the frequency at which they were primed. These results demonstrate a critical role for initial precursor frequency in determining the CD8+ T cell requirement for CD28- and CD154-mediated costimulatory signals during graft rejection.
Collapse
Affiliation(s)
- Mandy L Ford
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Garrod KR, Chang CK, Liu FC, Brennan TV, Foster RD, Kang SM. Targeted Lymphoid Homing of Dendritic Cells Is Required for Prolongation of Allograft Survival. THE JOURNAL OF IMMUNOLOGY 2006; 177:863-8. [DOI: 10.4049/jimmunol.177.2.863] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
35
|
Tang AL, Bingaman AW, Kadavil EA, Leeser DB, Farber DL. Generation and functional capacity of polyclonal alloantigen-specific memory CD4 T cells. Am J Transplant 2006; 6:1275-84. [PMID: 16686752 DOI: 10.1111/j.1600-6143.2006.01317.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alloreactive memory T cells can significantly impact graft survival due to their enhanced functional capacities, diverse tissue distribution and resistance to tolerance induction and depletional strategies. However, their role in allograft rejection is not well understood primarily due to the lack of suitable in vivo models. In this study, we use a novel approach to generate long-lived polyclonal alloreactive memory CD4 T cells from adoptive transfer of alloantigen-activated precursors into mouse hosts. We demonstrate that CD25 upregulation is a marker for precursors to alloantigen-specific memory and have created a new mouse model that features an expanded population of polyclonal alloreactive memory T cells that is distinguishable from the naive T-cell population. Furthermore, we show that alloreactive memory T cells exhibit rapid recall effector responses with predominant IFN-gamma and IL-2 production, and mediate vigorous allograft rejection. Interestingly, while we found a heterogeneous distribution of allomemory T cells in lymphoid and nonlymphoid tissues, they were all predominantly of the effector-memory (CD62Llo) phenotype. Our results present a unique model for the generation and tracking of polyclonal allospecific memory CD4 T cells in vivo and reveal insights into the distinct and robust nature of alloreactive T-cell memory.
Collapse
Affiliation(s)
- A L Tang
- Division of Transplantation, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | |
Collapse
|
36
|
Kapessidou Y, Habran C, Buonocore S, Flamand V, Barvais L, Goldman M, Braun MY. The replacement of graft endothelium by recipient-type cells conditions allograft rejection mediated by indirect pathway CD4(+) T cells. Transplantation 2006; 81:726-35. [PMID: 16534475 DOI: 10.1097/01.tp.0000184444.93108.d1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Whereas the participation of alloreactive T cells sensitized by indirect allorecognition in graft rejection is well documented, the nature of recipient antigen presenting cells recognized by indirect pathway CD4(+) T cells within the graft has yet to be identified. The purpose of this study was to determine the role played by graft endothelium replacement in the immune recognition of cardiac allografts rejected by indirect pathway CD4(+) T cells. METHODS Transgenic RAG2(-/-) mice expressing I-A(b)-restricted male antigen H-Y-specific TcR were studied for their capacity to reject H-2(k) male cardiac allografts. Chronic vascular rejection in this model was due to the indirect recognition of H-Y antigen shed from H-2(k) male allograft and presented by the recipient's own I-A(b) APC to transgenic T cells. RESULTS Immunohistochemical analysis of rejected grafts revealed the presence of numerous microvascular endothelial cells (EC) that expressed recipient's I-A MHC class II molecules. This observation suggested that graft endothelium replacement by I-A(b)-positive cells of recipient origin could stimulate the rejection of male H-2(k) graft by I-A(b)--restricted H-Y--specific T cells. To investigate further this possibility, hearts from H-2(b)--into--H-2(k) irradiation bone marrow (BM) chimera were transplanted in transgenic recipients. A direct correlation was observed between the presence of I-A(b)-positive EC within myocardial microvessels and the induction of acute rejection of chimeric H-2(k) male cardiac allografts transplanted in transgenic recipients. CONCLUSIONS We conclude that graft endothelium replacement by recipient-type cells is required for the rejection of cardiac allograft mediated by indirect pathway alloreactive CD4(+) T cells.
Collapse
Affiliation(s)
- Yota Kapessidou
- Institute for Medical Immunology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
37
|
Ochando JC, Homma C, Yang Y, Hidalgo A, Garin A, Tacke F, Angeli V, Li Y, Boros P, Ding Y, Jessberger R, Trinchieri G, Lira SA, Randolph GJ, Bromberg JS. Alloantigen-presenting plasmacytoid dendritic cells mediate tolerance to vascularized grafts. Nat Immunol 2006; 7:652-62. [PMID: 16633346 DOI: 10.1038/ni1333] [Citation(s) in RCA: 533] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Accepted: 03/10/2006] [Indexed: 02/05/2023]
Abstract
The induction of alloantigen-specific unresponsiveness remains an elusive goal in organ transplantation. Here we identify plasmacytoid dendritic cells (pDCs) as phagocytic antigen-presenting cells essential for tolerance to vascularized cardiac allografts. Tolerizing pDCs acquired alloantigen in the allograft and then moved through the blood to home to peripheral lymph nodes. In the lymph node, alloantigen-presenting pDCs induced the generation of CCR4+ CD4+ CD25+ Foxp3+ regulatory T cells (Treg cells). Depletion of pDCs or prevention of pDC lymph node homing inhibited peripheral Treg cell development and tolerance induction, whereas adoptive transfer of tolerized pDCs induced Treg cell development and prolonged graft survival. Thus, alloantigen-presenting pDCs home to the lymph nodes in tolerogenic conditions, where they mediate alloantigen-specific Treg cell development and allograft tolerance.
Collapse
Affiliation(s)
- Jordi C Ochando
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York 10029-6574, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Habicht A, Clarkson MR, Yang J, Henderson J, Brinkmann V, Fernandes S, Jurewicz M, Yuan X, Sayegh MH. Novel insights into the mechanism of action of FTY720 in a transgenic model of allograft rejection: implications for therapy of chronic rejection. THE JOURNAL OF IMMUNOLOGY 2006; 176:36-42. [PMID: 16365393 DOI: 10.4049/jimmunol.176.1.36] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
FTY720 is a high-affinity agonist at the sphingosine 1-phosphate receptor 1 that prevents lymphocyte egress from lymphoid tissue and prolongs allograft survival in several animal models of solid organ transplantation. In this study we used a recently developed adoptive transfer model of TCR transgenic T cells to track allospecific CD4+ T cell expansion and trafficking characteristics, cytokine secretion profiles, and surface phenotype in vivo in the setting of FTY720 administration. We report that FTY720 administration had no effect on alloantigen-driven T cell activation, proliferation, acquisition of effector-memory function, or T cell apoptosis. However, FTY720 caused a reversible sequestration of alloantigen-specific effector-memory T cells in regional lymphoid tissue associated with a decrease in T cell infiltration within the allograft and a subsequent prolongation in allograft survival. Furthermore, delayed administration of FTY720 in a cardiac model of chronic allograft rejection attenuated the progression of vasculopathy and tissue fibrosis consistent with the hypothesis that FTY720 interrupts the trafficking of activated effector-memory T cells. These data have important implications for targeting the sphingosine 1-phosphate receptor 1 in solid organ transplantation.
Collapse
Affiliation(s)
- Antje Habicht
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sánchez-Fueyo A, Sandner S, Habicht A, Mariat C, Kenny J, Degauque N, Zheng XX, Strom TB, Turka LA, Sayegh MH. Specificity of CD4+CD25+ regulatory T cell function in alloimmunity. THE JOURNAL OF IMMUNOLOGY 2006; 176:329-34. [PMID: 16365425 PMCID: PMC2841032 DOI: 10.4049/jimmunol.176.1.329] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD4+CD25+ regulatory T cells (TRegs) are critical for the acquisition of peripheral allograft tolerance. However, it is unclear whether TRegs are capable of mediating alloantigen-specific suppressive effects and, hence, contributing to the specificity of the tolerant state. In the current report we have used the ABM TCR transgenic (Tg) system, a C57BL/6-derived strain in which CD4+ T cells directly recognize the allogeneic MHC-II molecule I-A(bm12), to assess the capacity of TRegs to mediate allospecific effects. In these mice, 5-6% of Tg CD4+ T cells exhibit conventional markers of the TReg phenotype. ABM TRegs are more effective than wild-type polyclonal TRegs at suppressing effector immune responses directed against I-A(bm12) alloantigen both in vitro and in vivo. In contrast, they are incapable of suppressing responses directed against third-party alloantigens unless these are expressed in the same allograft as I-A(bm12). Taken together, our results indicate that in transplantation, TReg function is dependent on TCR stimulation, providing definitive evidence for their specificity in the regulation of alloimmune responses.
Collapse
Affiliation(s)
- Alberto Sánchez-Fueyo
- Department of Medicine and Department of Surgery, Transplant Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Sigrid Sandner
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| | - Antje Habicht
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| | - Christophe Mariat
- Department of Medicine and Department of Surgery, Transplant Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - James Kenny
- Department of Medicine and Department of Surgery, Transplant Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Nicolas Degauque
- Department of Medicine and Department of Surgery, Transplant Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Xin Xiao Zheng
- Department of Medicine and Department of Surgery, Transplant Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Terry B. Strom
- Department of Medicine and Department of Surgery, Transplant Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | | | - Mohamed H. Sayegh
- Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
- Address correspondence and reprint requests to Dr. Mohamed H. Sayegh, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115.
| |
Collapse
|
40
|
Kashizuka H, Sho M, Nomi T, Ikeda N, Kuzumoto Y, Akashi S, Tsurui Y, Mizuno T, Kanehiro H, Yagita H, Nakajima Y, Sayegh MH. Role of the ICOS-B7h Costimulatory Pathway in the Pathophysiology of Chronic Allograft Rejection. Transplantation 2005; 79:1045-50. [PMID: 15880041 DOI: 10.1097/01.tp.0000161665.35243.21] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Inducible costimulator (ICOS) is the third member of the CD28 superfamily and has a unique role in T cell activation and function. Recent studies indicated that the ICOS-B7h pathway plays an important role in alloimmune responses. We further investigated the role of the ICOS pathway in the pathologic process of chronic rejection in vivo. METHODS An established major histocompatibility complex class II disparate cardiac transplantation model was used. We treated mice with a blocking anti-B7h monoclonal antibody (mAb) either in the initiation phase (early blockade) or in the progression phase (delayed blockade) of disease. In addition, some mice received mAb in the entire period (whole blockade). At 6 weeks after transplantation, cardiac grafts were evaluated by histopathologic analysis in terms of vasculopathy, fibrosis, and cellular infiltration. The intragraft expressions of cytokines and chemokines were also examined by quantitative real-time polymerase chain reaction analysis. RESULTS Early blockade of the ICOS-B7h pathway did not show any protective effect on chronic allograft rejection compared with untreated controls. In contrast, delayed blockade significantly inhibited the development of vasculopathy, fibrosis, and cellular infiltration (P=0.043, P=0.004, and P=0.03 vs. untreated control, respectively). Interestingly, whole blockade did not prevent the chronic rejection process. Furthermore, the inhibitory effect of delayed ICOS blockade on chronic rejection was associated with down-regulation of local intragraft expression of several cytokines and chemokines. CONCLUSIONS These data suggest that the ICOS-B7h pathway is critical in the activation of effector/memory T cells that are necessary for the progression of chronic rejection and provide the rationale to develop novel and specific therapies to prevent this process.
Collapse
Affiliation(s)
- Hisanori Kashizuka
- Department of Surgery, Nara Medical University School of Medicine, Kashihara, Nara, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Benghiat FS, Graca L, Braun MY, Detienne S, Moore F, Buonocore S, Flamand V, Waldmann H, Goldman M, Le Moine A. Critical influence of natural regulatory CD25+ T cells on the fate of allografts in the absence of immunosuppression. Transplantation 2005; 79:648-54. [PMID: 15785370 DOI: 10.1097/01.tp.0000155179.61445.78] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Allografts are occasionally accepted in the absence of immunosuppression. Because naturally occurring CD4(+)CD25(+) regulatory T cells (natural CD25(+) Treg cells) have been shown to inhibit allograft rejection, we investigated their influence on the outcome of allografts in nonimmunosuppressed mouse recipients. METHODS We compared survival times of male CBA/Ca skin grafts in female CBA/Ca recipients expressing a transgenic anti-HY T-cell receptor on a RAG-1(+/+) (A1[M]RAG+) or a RAG-1(-/-) (A1[M]RAG-) background. Depletion of natural CD25(+) Treg cells in A1[M]RAG+ mice was achieved by in vivo administration of the PC61 monoclonal antibody. The influence of natural CD25(+) Treg cells on the fate of major histocompatibility complex class II-mismatched (C57BL/6X bm12)F1 skin or bm12 heart transplants in C57BL/6 recipients was also assessed. Finally, we investigated the impact of natural CD25(+) Treg cells on the production of T-helper (Th)1 and Th2 cytokines in mixed lymphocyte cultures between C57BL/6 CD4(+) CD25(-) T cells as responders and bm12 or (C57BL/6X bm12)F1 antigen-presenting cells as stimulators. RESULTS Male allografts were spontaneously accepted by female A1(M)RAG+ mice but readily rejected by female A1(M)RAG+ mice depleted of natural CD25(+) Treg cells by pretreatment with the PC61 monoclonal antibody. Depletion of CD25(+) Treg cells also enhanced eosinophil-determined rejection of (C57BL/6X bm12)F1 skin grafts or bm12 cardiac grafts in C57BL/6 recipients. Finally, natural CD25(+) Treg cells inhibited the production of interleukin (IL)-2, interferon-gamma, IL-5, and IL-13 in mixed lymphocyte culture in a dose-dependent manner. CONCLUSION Natural CD25(+) Treg cells control Th1- and Th2-type allohelper T-cell responses and thereby influence the fate of allografts in nonimmunosuppressed recipients.
Collapse
MESH Headings
- Animals
- Cytokines/immunology
- Cytokines/metabolism
- Female
- Graft Rejection/immunology
- Graft Survival/immunology
- Heart Transplantation/immunology
- Heart Transplantation/pathology
- Immunosuppression Therapy
- Lymphocyte Culture Test, Mixed
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Rats
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Interleukin-2/immunology
- Receptors, Interleukin-2/metabolism
- Skin Transplantation/immunology
- Skin Transplantation/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Transplantation
- Transplantation, Homologous/immunology
Collapse
Affiliation(s)
- Fleur Samantha Benghiat
- Institute for Medical Immunology, Université Libre de Bruxelles, 8, rue Adrienne Bolland, B-6041 Charleroi (Gosselies), Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sandner SE, Clarkson MR, Salama AD, Sanchez-Fueyo A, Domenig C, Habicht A, Najafian N, Yagita H, Azuma M, Turka LA, Sayegh MH. Role of the Programmed Death-1 Pathway in Regulation of Alloimmune Responses In Vivo. THE JOURNAL OF IMMUNOLOGY 2005; 174:3408-15. [PMID: 15749874 DOI: 10.4049/jimmunol.174.6.3408] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Programmed death-1 (PD-1), an inhibitory receptor up-regulated on activated T cells, has been shown to play a critical immunoregulatory role in peripheral tolerance, but its role in alloimmune responses is poorly understood. Using a novel alloreactive TCR-transgenic model system, we examined the functions of this pathway in the regulation of alloreactive CD4+ T cell responses in vivo. PD-L1, but not PD-1 or PD-L2, blockade accelerated MHC class II-mismatched skin graft (bm12 (I-Abm12) into B6 (I-Ab)) rejection in a similar manner to CTLA-4 blockade. In an adoptive transfer model system using the recently described anti-bm12 (ABM) TCR-transgenic mice directly reactive to I-Abm12, PD-1 and PD-L1 blockade enhanced T cell proliferation early in the immune response. In contrast, at a later time point preceding accelerated allograft rejection, only PD-L1 blockade enhanced T cell proliferation. In addition, PD-L1 blockade enhanced alloreactive Th1 cell differentiation. Apoptosis of alloantigen-specific T cells was inhibited significantly by PD-L1 but not PD-1 blockade, indicating that PD-1 may not be the receptor for the apoptotic effect of the PD-L1-signaling pathway. Interestingly, the effect of PD-L1 blockade was dependent on the presence of CD4+ CD25+ regulatory T cells in vivo. These data demonstrate a critical role for the PD-1 pathway, particularly PD-1/PD-L1 interactions, in the regulation of alloimmune responses in vivo.
Collapse
Affiliation(s)
- Sigrid E Sandner
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yamada A, Salama AD, Sho M, Najafian N, Ito T, Forman JP, Kewalramani R, Sandner S, Harada H, Clarkson MR, Mandelbrot DA, Sharpe AH, Oshima H, Yagita H, Chalasani G, Lakkis FG, Auchincloss H, Sayegh MH. CD70 Signaling Is Critical for CD28-Independent CD8+T Cell-Mediated Alloimmune Responses In Vivo. THE JOURNAL OF IMMUNOLOGY 2005; 174:1357-64. [PMID: 15661893 DOI: 10.4049/jimmunol.174.3.1357] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The inability to reproducibly induce robust and durable transplant tolerance using CD28-B7 pathway blockade is in part related to the persistence of alloreactive effector/memory CD8(+) T cells that are less dependent on this pathway for their cellular activation. We studied the role of the novel T cell costimulatory pathway, CD27-CD70, in alloimmunity in the presence and absence of CD28-B7 signaling. CD70 blockade prolonged survival of fully mismatched vascularized cardiac allografts in wild-type murine recipients, and in CD28-deficient mice induced long-term survival while significantly preventing the development of chronic allograft vasculopathy. CD70 blockade had little effect on CD4(+) T cell function but prevented CD8(+) T cell-mediated rejection, inhibited the proliferation and activation of effector CD8(+) T cells, and diminished the expansion of effector and memory CD8(+) T cells in vivo. Thus, the CD27-CD70 pathway is critical for CD28-independent effector/memory CD8(+) alloreactive T cell activation in vivo. These novel findings have important implications for the development of transplantation tolerance-inducing strategies in primates and humans, in which CD8(+) T cell depletion is currently mandatory.
Collapse
MESH Headings
- Acute Disease
- Adoptive Transfer
- Animals
- Antibodies, Blocking/administration & dosage
- Antibodies, Monoclonal/administration & dosage
- Antigens, CD/immunology
- Antigens, CD/physiology
- CD27 Ligand
- CD28 Antigens/genetics
- CD28 Antigens/physiology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/transplantation
- CD8-Positive T-Lymphocytes/immunology
- Chronic Disease
- Down-Regulation/immunology
- Graft Rejection/genetics
- Graft Rejection/immunology
- Graft Survival/genetics
- Graft Survival/immunology
- Heart Transplantation/immunology
- Immunologic Memory/immunology
- Isoantibodies/biosynthesis
- Isoantibodies/blood
- Killer Cells, Natural/immunology
- Lymphocyte Activation/immunology
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/immunology
- Membrane Proteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Signal Transduction/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Transplantation, Heterotopic/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Akira Yamada
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sandner SE, Clarkson MR, Salama AD, Sanchez-Fueyo A, Yagita H, Turka LA, Sayegh MH. Mechanisms of tolerance induced by donor-specific transfusion and ICOS-B7h blockade in a model of CD4+ T-cell-mediated allograft rejection. Am J Transplant 2005; 5:31-9. [PMID: 15636609 DOI: 10.1111/j.1600-6143.2004.00640.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The inducible co-stimulatory molecule (ICOS) has been shown to play a critical role in T-cell activation and differentiation, and the regulation of alloimmune responses in vivo. Using an MHC class II mismatched model of CD4(+) T-cell-mediated rejection, we found that treatment of mice with DST and ICOS-B7h blockade induced long-term skin allograft survival and donor-specific transplantation tolerance. ICOS blockade, either during antigen priming or during the effector phase, previously shown to alter the outcome of the immune response, had a similar effect on graft survival. DST and anti-B7h mAb reduced the frequency of IFN-gamma-producing allospecific cells but did not produce deviation to a T(H)2 phenotype. In an adoptive transfer model using ABM TCR transgenic mice directly reactive to I-A(bm12), DST and anti-B7h mAb reduced the number of allospecific CD4(+) T cells and increased CD4(+) T-cell apoptosis. These data demonstrate that DST and anti-B7h mAb induces transplantation tolerance to MHC class II mismatched skin grafts by a reduction of the alloreactive clone size that is, at least in part, dependent on apoptosis of host alloantigen-specific CD4(+) T cells.
Collapse
Affiliation(s)
- Sigrid E Sandner
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Honjo K, Xu XY, Kapp JA, Bucy RP. Activation and migration of allo-peptide specific TCR transgenic T cells in cardiac allograft rejection. Cell Immunol 2004; 230:44-55. [PMID: 15541718 DOI: 10.1016/j.cellimm.2004.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2004] [Accepted: 09/03/2004] [Indexed: 11/24/2022]
Abstract
T cells from TCR transgenic mice, expressing receptors specific for an allogeneic MHC class I peptide, were used to track T cell activation and migration in normal adoptive recipients that were subsequently transplanted with heterotopic hearts that were syngeneic except for a transgenic MHC class I antigen. T cells rapidly disappeared from the blood into the lymphoid tissues where they were activated within one day after transplantation. T cells initially formed discrete clusters in the spleen and lymph nodes. After proliferating for 2-4 days in lymphoid tissues, T cells reappeared in the blood and migrated to the heart and the intestines. The T cells underwent another round of proliferation in the heart, but not the intestines, and induced cardiac rejection uniformly on 6 day.
Collapse
Affiliation(s)
- Kazuhito Honjo
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233-7331, USA
| | | | | | | |
Collapse
|
46
|
Honjo K, Xu XY, Bucy RP. CD4+ T-cell receptor transgenic T cells alone can reject vascularized heart transplants through the indirect pathway of alloantigen recognition. Transplantation 2004; 77:452-5. [PMID: 14966425 DOI: 10.1097/01.tp.0000112937.12491.42] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The vast array of epitopes presented by allografts and the diversity of T cells responding to them complicates mechanistic studies of rejection. To minimize these problems, we developed a transgenic (Tg) model system limited to a single T-cell receptor (TCR)/peptide/major histocompatibility complex molecule. Two alloantigen-specific CD4 T-cell clones were used to isolate cDNA encoding the TCRalpha and TCRbeta chains that recognize the Kd54-68/I-Ab epitope. Two different TCR Tg lines were produced in C57BL/6 (B6) mice and crossed onto the B6.Rag1-/- background. B6.Rag1-/- recipients of T cells from TCR Tg Rag1-/-mice promptly rejected B10.D2, but not irrelevant B10.BR, cardiac grafts. Thus, a single allogeneic epitope presented by self-major histocompatibility complex class II is sufficient to activate TCR Tg T cells and serve as a target for rejection.
Collapse
Affiliation(s)
- Kazuhito Honjo
- Department of Pathology, University of Alabama at Birmingham, AL 65294-2170, USA
| | | | | |
Collapse
|
47
|
Abstract
Allograft arteriopathy is a vascular intimal proliferative process that occurs in all solid organ transplants and stands as the single most significant obstacle to successful long-term solid organ transplantation; it shares a number of pathologic features with restenosis lesions and atherosclerosis. This article will review some of the newer developments in our understanding of the immunological and vascular biology underpinnings of the disease, including the roles played by cytokine and chemokine mediators in recruiting and activating both inflammatory cells, as well as smooth muscle cell precursors.
Collapse
Affiliation(s)
- Richard N Mitchell
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur/NRB 730D, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Sandner SE, Salama AD, Houser SL, Palmer E, Turka LA, Sayegh MH. New TCR transgenic model for tracking allospecific CD4 T-cell activation and tolerance in vivo. Am J Transplant 2003; 3:1242-50. [PMID: 14510697 DOI: 10.1046/j.1600-6143.2003.00220.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We have developed an adoptive transfer model system to visualize the dynamics of alloantigen-specific CD4+ T-cell activation in vivo. Using TCR-transgenic (tg) mice reactive to I-Ab(m12), we studied the clonal expansion and differentiation of alloreactive T cells by tracking the fate of adoptively transferred TCR-tg CD4+ T cells in syngeneic mice transplanted with skin grafts expressing I-Ab(m12). Following transplantation, alloantigen-specific TCR-tg CD4+ T-cell expansion was observed initially in the draining lymph nodes followed by the spleen. TCR-tg CD4+ T cells up-regulated CD69 and CD25 expression, developed an effector/memory surface phenotype and produced IFN-gamma in response to alloantigen ex vivo. Furthermore, we validate the model system as a means for studying the effects of tolerogenic regimens on alloreactive CD4+ T cells, demonstrating that CTLA4Ig inhibits alloantigen-dependent clonal expansion and effector function of TCR-tg CD4+ T cells in vivo. We describe the first model for tracking alloreactive CD4+ T-cell activation in vivo. It provides a powerful tool for studying CD4+ T-cell mediated alloimmune responses and mechanisms of tolerance induction in vivo.
Collapse
Affiliation(s)
- Sigrid E Sandner
- Division of Nephrology, The Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
49
|
Harada H, Salama AD, Sho M, Izawa A, Sandner SE, Ito T, Akiba H, Yagita H, Sharpe AH, Freeman GJ, Sayegh MH. The role of the ICOS-B7h T cell costimulatory pathway in transplantation immunity. J Clin Invest 2003. [DOI: 10.1172/jci200317008] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
50
|
Harada H, Salama AD, Sho M, Izawa A, Sandner SE, Ito T, Akiba H, Yagita H, Sharpe AH, Freeman GJ, Sayegh MH. The role of the ICOS-B7h T cell costimulatory pathway in transplantation immunity. J Clin Invest 2003; 112:234-43. [PMID: 12865411 PMCID: PMC164288 DOI: 10.1172/jci17008] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2002] [Accepted: 04/22/2003] [Indexed: 11/17/2022] Open
Abstract
Inducible costimulatory molecule (ICOS) plays a pivotal role in T cell activation and Th1/Th2 differentiation. ICOS blockade has disparate effects on immune responses depending on the timing of blockade. Its role in transplantation immunity, however, remains incompletely defined. We used a vascularized mouse cardiac allograft model to explore the role of ICOS signaling at different time points after transplantation, targeting immune initiation (early blockade) or the immune effector phase (delayed blockade). In major histocompatibility-mismatched recipients, ICOS blockade prolonged allograft survival using both protocols but did so more effectively in the delayed-treatment group. By contrast, in minor histocompatibility-mismatched recipients, early blockade accelerated rejection and delayed blockade prolonged graft survival. Alloreactive CD4+ T cell expansion and alloantibody production were suppressed in both treatment groups, whereas only delayed blockade resulted in suppression of effector CD8+ T cell generation. After delayed ICOS blockade, there was a diminished frequency of allospecific IL-10-producing cells and an increased frequency of both IFN-gamma- and IL-4-producing cells. The beneficial effects of ICOS blockade in regulating allograft rejection were seen in the absence of CD28 costimulation but required CD8+ cells, cytotoxic T lymphocyte antigen-4, and an intact signal transducer and activator of transcription-6 pathway. These data define the complex functions of the ICOS-B7h pathway in regulating alloimmune responses in vivo.
Collapse
Affiliation(s)
- Hiroshi Harada
- Department of Medicine, Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|