1
|
Wragg KM, Worley MJ, Deng JC, Salmon M, Goldstein DR. Deficiency in the mitophagy mediator Parkin accelerates murine skin allograft rejection. Am J Transplant 2024; 24:2174-2186. [PMID: 39142471 PMCID: PMC11588513 DOI: 10.1016/j.ajt.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/21/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
Alterations in mitochondrial function and associated quality control programs, including mitochondrial-specific autophagy, termed mitophagy, are gaining increasing recognition in the context of disease. However, the role of mitophagy in organ transplant rejection remains poorly understood. Using mice deficient in Parkin, a ubiquitin ligase that tags damaged or dysfunctional mitochondria for autophagic clearance, we assessed the impact of Parkin-dependent mitophagy on skin-graft rejection. We observed accelerated graft loss in Parkin-deficient mice across multiple skin graft models. Immune cell distributions posttransplant were largely unperturbed compared to wild-type; however, the CD8+ T cells of Parkin-deficient mice expressed more T-bet, IFNγ, and Ki67, indicating greater priming toward effector function. This was accompanied by increased circulating levels of IL-12p70 in Parkin-deficient mice. Using a mixed leukocyte reaction, we demonstrated that naïve Parkin-deficient CD4+ and CD8+ T cells exhibit enhanced activation marker expression and proliferative responses to alloantigen, which were attenuated with administration of a pharmacological mitophagy inducer (p62-mediated mitophagy inducer), known to increase mitophagy in the absence of a functional PINK1-Parkin pathway. These findings indicate a role for Parkin-dependent mitophagy in curtailing skin-graft rejection.
Collapse
Affiliation(s)
- Kathleen M Wragg
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew J Worley
- Pulmonary Division, University of Michigan, Ann Arbor, Michigan, USA
| | - Jane C Deng
- Pulmonary Division, University of Michigan, Ann Arbor, Michigan, USA; Veterans Affairs Ann Arbor, Ann Arbor, Michigan, USA
| | - Morgan Salmon
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan, USA; Frankel Cardiovascular Center, University of Michigan School of Medicine, Ann Arbor, Michigan, USA.
| | - Daniel R Goldstein
- Frankel Cardiovascular Center, University of Michigan School of Medicine, Ann Arbor, Michigan, USA; Department of Medicine, Cardiology Division, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
2
|
Nicosia M, Valujskikh A. Recognizing Complexity of CD8 T Cells in Transplantation. Transplantation 2024; 108:2186-2196. [PMID: 38637929 PMCID: PMC11489323 DOI: 10.1097/tp.0000000000005001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
The major role of CD8 + T cells in clinical and experimental transplantation is well documented and acknowledged. Nevertheless, the precise impact of CD8 + T cells on graft tissue injury is not completely understood, thus impeding the development of specific treatment strategies. The goal of this overview is to consider the biology and functions of CD8 + T cells in the context of experimental and clinical allotransplantation, with special emphasis on how this cell subset is affected by currently available and emerging therapies.
Collapse
Affiliation(s)
- Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
3
|
Laws LH, Parker CE, Cherala G, Koguchi Y, Waisman A, Slifka MK, Oberbarnscheidt MH, Obhrai JS, Yeung MY, Riella LV. Inflammation Causes Resistance to Anti-CD20-Mediated B Cell Depletion. Am J Transplant 2016; 16:3139-3149. [PMID: 27265023 PMCID: PMC5334788 DOI: 10.1111/ajt.13902] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/11/2016] [Accepted: 05/23/2016] [Indexed: 01/25/2023]
Abstract
B cells play a central role in antibody-mediated rejection and certain autoimmune diseases. However, B cell-targeted therapy such as anti-CD20 B cell-depleting antibody (aCD20) has yielded mixed results in improving outcomes. In this study, we investigated whether an accelerated B cell reconstitution leading to aCD20 depletion resistance could account for these discrepancies. Using a transplantation model, we found that antigen-independent inflammation, likely through toll-like receptor (TLR) signaling, was sufficient to mitigate B cell depletion. Secondary lymphoid organs had a quicker recovery of B cells when compared to peripheral blood. Inflammation altered the pharmacokinetics (PK) and pharmacodynamics (PD) of aCD20 therapy by shortening drug half-life and accelerating the reconstitution of the peripheral B cell pool by bone marrow-derived B cell precursors. IVIG (intravenous immunoglobulin) coadministration also shortened aCD20 drug half-life and led to accelerated B cell recovery. Repeated aCD20 dosing restored B cell depletion and delayed allograft rejection, especially B cell-dependent, antibody-independent allograft rejection. These data demonstrate the importance of further clinical studies of the PK/PD of monoclonal antibody treatment in inflammatory conditions. The data also highlight the disconnect between B cell depletion on peripheral blood compared to secondary lymphoid organs, the deleterious effect of IVIG when given with aCD20 and the relevance of redosing of aCD20 for effective B cell depletion in alloimmunity.
Collapse
Affiliation(s)
| | | | - Ganesh Cherala
- Department of Pharmacy Practice, College of Pharmacy, Oregon State University
| | - Yoshinobu Koguchi
- Molecular Microbiology & Immunology, Oregon Health & Science University
| | - Ari Waisman
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz
| | - Mark K. Slifka
- Oregon National Primate Research Center, Oregon Health & Science University
| | | | | | - Melissa Y. Yeung
- Schuster Transplant Research Center, Renal Division, Brigham & Women's Hospital, Harvard Medical School
| | - Leonardo V. Riella
- Schuster Transplant Research Center, Renal Division, Brigham & Women's Hospital, Harvard Medical School
| |
Collapse
|
4
|
Casiraghi F, Perico N, Cortinovis M, Remuzzi G. Mesenchymal stromal cells in renal transplantation: opportunities and challenges. Nat Rev Nephrol 2016; 12:241-53. [DOI: 10.1038/nrneph.2016.7] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
5
|
Holvoet B, Quattrocelli M, Belderbos S, Pollaris L, Wolfs E, Gheysens O, Gijsbers R, Vanoirbeek J, Verfaillie CM, Sampaolesi M, Deroose CM. Sodium Iodide Symporter PET and BLI Noninvasively Reveal Mesoangioblast Survival in Dystrophic Mice. Stem Cell Reports 2015; 5:1183-1195. [PMID: 26626179 PMCID: PMC4682284 DOI: 10.1016/j.stemcr.2015.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/29/2015] [Accepted: 10/29/2015] [Indexed: 01/27/2023] Open
Abstract
Muscular dystrophies are a heterogeneous group of myopathies, characterized by muscle weakness and degeneration, without curative treatment. Mesoangioblasts (MABs) have been proposed as a potential regenerative therapy. To improve our understanding of the in vivo behavior of MABs and the effect of different immunosuppressive therapies, like cyclosporine A or co-stimulation-adhesion blockade therapy, on cell survival noninvasive cell monitoring is required. Therefore, cells were transduced with a lentiviral vector encoding firefly luciferase (Fluc) and the human sodium iodide transporter (hNIS) to allow cell monitoring via bioluminescence imaging (BLI) and small-animal positron emission tomography (PET). Non-H2 matched mMABs were injected in the femoral artery of dystrophic mice and were clearly visible via small-animal PET and BLI. Based on noninvasive imaging data, we were able to show that co-stim was clearly superior to CsA in reducing cell rejection and this was mediated via a reduction in cytotoxic T cells and upregulation of regulatory T cells. Longitudinal monitoring of murine mesoangioblasts with BLI and small-animal PET Noninvasive evaluation of immune suppressant efficacy Inhibition of co-stimulation outperformed cyclosporin Inhibition of co-stimulation reduced cytotoxic and upregulated regulatory T cells
Collapse
Affiliation(s)
- Bryan Holvoet
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven 3000, Belgium
| | - Mattia Quattrocelli
- Department of Development and Regeneration, Translational Cardiomyology Lab, KU Leuven, Leuven 3000, Belgium
| | - Sarah Belderbos
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven 3000, Belgium
| | - Lore Pollaris
- Department of Public Health and Primary Care, Centre for Environment and Health, KU Leuven, Leuven 3000, Belgium
| | - Esther Wolfs
- Department of Morphology, Biomedical Research Institute, Lab of Histology, Universiteit Hasselt, Diepenbeek 3590, Belgium
| | - Olivier Gheysens
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven 3000, Belgium
| | - Rik Gijsbers
- Department of Pharmaceutical and Pharmacological Sciences, Laboratory of Molecular Virology and Gene Therapy, Leuven Viral Vector Core, KU Leuven, Leuven 3000, Belgium
| | - Jeroen Vanoirbeek
- Department of Public Health and Primary Care, Centre for Environment and Health, KU Leuven, Leuven 3000, Belgium
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Leuven 3000, Belgium
| | - Maurilio Sampaolesi
- Department of Development and Regeneration, Translational Cardiomyology Lab, KU Leuven, Leuven 3000, Belgium
| | - Christophe M Deroose
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven 3000, Belgium.
| |
Collapse
|
6
|
Shibasaki Y, Matsuura Y, Toda H, Imabayashi N, Nishino T, Uzumaki K, Hatanaka C, Yabu T, Moritomo T, Nakanishi T. Kinetics of lymphocyte subpopulations in allogeneic grafted scales of ginbuna crucian carp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 52:75-80. [PMID: 25917429 DOI: 10.1016/j.dci.2015.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 04/19/2015] [Accepted: 04/20/2015] [Indexed: 06/04/2023]
Abstract
In mammals the rejection of allografts is primarily accomplished by cell-mediated immunity including T cells. Recently, considerable studies reveal the existence of helper and cytotoxic T cell subsets in fish. Here we investigate the kinetics of CD4(+) and CD8α(+) T cells along with sIgM(+) cells and phagocytic cells in an allogeneic scale graft model using ginbuna crucian carp for understanding the mechanisms of cell-mediated immune response. The results showed that CD4(+) T cells first infiltrated into allogeneic scales followed by CD8α(+) and sIgM(+) cells, and finally phagocytic cells appeared in the graft. Furthermore, most of the CD8α(+) T cells appeared on the border of the allografted scales at the time of rejection. These results suggest that T cells play crucial roles and work together with other cell types for completion of allograft rejection.
Collapse
Affiliation(s)
- Yasuhiro Shibasaki
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Yuta Matsuura
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Hideaki Toda
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Nozomi Imabayashi
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Tatsuyuki Nishino
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Kosuke Uzumaki
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Chihiro Hatanaka
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Takeshi Yabu
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Tadaaki Moritomo
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| | - Teruyuki Nakanishi
- Department of Veterinary Medicine, Nihon University, Fujisawa, Kanagawa 252-8510, Japan.
| |
Collapse
|
7
|
Recognition of Foreign Antigen and Foreign Major Histocompatibility Complex. Xenotransplantation 2014. [DOI: 10.1128/9781555818043.ch9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
8
|
Abstract
The most important transplantation antigens in the discrimination between "self" and "nonself" are encoded by genes in the major histocompatibility complex (MHC) locus (H-2 in mice). It has been assumed that T lymphocytes are the effector cells for allograft rejection, as athymic nude rodents fail to reject allografts. In 1988, we i.p. transplanted Meth A (H-2D(d)K(d)) tumor cells into C57BL/6 (H-2D(b)K(b)) mice and found macrophages to be cytotoxic against the allografts. In 1996, several groups using CD4 or CD8 knockout mice reported that non-T cells were the effector cells for the rejection of skin or organ allografts. In 1998, we ascertained that macrophages were the effector cells of skin allograft rejection. Recently, we isolated cDNA clones encoding monocyte/macrophage MHC receptors (MMRs) for H-2D(d) and H-2K(d); established H-2D(d)- and/or H-2K(d)-transgenic mice and lymphoma cells; and found, using MMR-deficient mice, that MMR and T-cell receptor were essential for the rejection of transgenic skin and lymphoma, respectively.
Collapse
Affiliation(s)
- Ryotaro Yoshida
- Department of Research Laboratory, Osaka Medical College, Takatsuki, Japan.
| |
Collapse
|
9
|
McFarland HI, Puig M, Grajkowska LT, Tsuji K, Lee JP, Mason KP, Verthelyi D, Rosenberg AS. Regulatory T cells in γ irradiation-induced immune suppression. PLoS One 2012; 7:e39092. [PMID: 22723935 PMCID: PMC3378522 DOI: 10.1371/journal.pone.0039092] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 05/18/2012] [Indexed: 11/24/2022] Open
Abstract
Sublethal total body γ irradiation (TBI) of mammals causes generalized immunosuppression, in part by induction of lymphocyte apoptosis. Here, we provide evidence that a part of this immune suppression may be attributable to dysfunction of immune regulation. We investigated the effects of sublethal TBI on T cell memory responses to gain insight into the potential for loss of vaccine immunity following such exposure. We show that in mice primed to an MHC class I alloantigen, the accelerated graft rejection T memory response is specifically lost several weeks following TBI, whereas identically treated naïve mice at the same time point had completely recovered normal rejection kinetics. Depletion in vivo with anti-CD4 or anti-CD25 showed that the mechanism involved cells consistent with a regulatory T cell (T reg) phenotype. The loss of the T memory response following TBI was associated with a relative increase of CD4+CD25+ Foxp3+ expressing T regs, as compared to the CD8+ T effector cells requisite for skin graft rejection. The radiation-induced T memory suppression was shown to be antigen-specific in that a third party ipsilateral graft rejected with normal kinetics. Remarkably, following the eventual rejection of the first MHC class I disparate skin graft, the suppressive environment was maintained, with markedly prolonged survival of a second identical allograft. These findings have potential importance as regards the immunologic status of T memory responses in victims of ionizing radiation exposure and apoptosis-inducing therapies.
Collapse
Affiliation(s)
- Hugh I McFarland
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Rahimpour A, Mattarollo SR, Yong M, Leggatt GR, Steptoe RJ, Frazer IH. γδ T cells augment rejection of skin grafts by enhancing cross-priming of CD8 T cells to skin-derived antigen. J Invest Dermatol 2012; 132:1656-64. [PMID: 22358058 PMCID: PMC3352982 DOI: 10.1038/jid.2012.16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gamma delta T cells (γδ T cells) possess innate-like properties and are proposed to bridge the gap between innate and adaptive immunity. In this study, we explored the role of γδ T cells in cutaneous immunity using a skin transplantation model. Following engraftment of skin expressing cell-associated model antigen (Ag) (ovalbumin) in epithelial keratinocytes, skin-resident γδ T cells enhanced graft rejection. Although the effector function of CD8 T cells was intact in the absence of γδ T cells, cross-priming of CD8 T cell to graft-derived Ag was impaired in the absence of γδ T cells. The reduced graft rejection and graft priming of γδ T-cell-deficient mice was evident in both acutely inflamed and well-healed grafting models. Furthermore, expression of the CD40 activation marker on migrating dendritic cells was lower in TCRδ(-/-) mice compared with wild-type mice, regardless of the presence or absence of inflammation associated with grafting. These results indicate that γδ T cells enhance graft priming and consequently the likelihood of a successful immune outcome in the context of skin graft rejection, suggesting that γδ T cells may be an important component of immunity to epithelial cancers or infection.
Collapse
Affiliation(s)
- Azad Rahimpour
- The University of Queensland Diamantina Institute, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
Gradually improved immunosuppression has contributed significantly to the progress achieved in transplantation medicine so far. Nevertheless, current drug regimens are associated with late graft loss--in particular as a result of immunologic damage or drug toxicity--and substantial morbidity. Recently, the costimulation blocker belatacept (marketed under the name Nulojix®) has been approved for immunosuppression in renal transplantation. Belatacept (a mutated version of CTLA4Ig) is a fusion protein rationally designed to block CD28, a critical activating receptor on T cells, by binding and saturating its ligands B7-1 and B7-2. In phase II and III trials, belatacept was compared with cyclosporine (in combination with basiliximab, MMF, and steroids). Advantages observed with belatacept include superior graft function, preservation of renal structure and improved cardiovascular risk profile. Concerns associated with belatacept are a higher frequency of cellular rejection episodes and more post-transplant lymphoproliferative disorder (PTLD) cases especially in EBV seronegative patients, who should be excluded from belatacept-based regimens. Thus, after almost three decades of calcineurin inhibitors as mainstay of immunosuppression, belatacept offers a potential alternative. In this article, we will provide an overview of belatacept's preclinical development and will discuss the available evidence from clinical trials.
Collapse
Affiliation(s)
- Thomas Wekerle
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria.
| | | |
Collapse
|
12
|
Robertson DM, Kalangara JP, Baucom RB, Petroll WM, Cavanagh HD. A reconstituted telomerase-immortalized human corneal epithelium in vivo: a pilot study. Curr Eye Res 2011; 36:706-12. [PMID: 21780919 DOI: 10.3109/02713683.2011.582662] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE Telomerase-immortalized human corneal epithelial cells have been reported to stratify and differentiate in vitro similar to native tissue. The purpose of this study was to assess the ability of a telomerase-immortalized human corneal epithelial cell line to generate a full thickness epithelium in vivo in athymic mice. METHODS Telomerized corneal epithelial cells were transduced with a retroviral vector encoding the herpes simplex thymidine kinase gene. Efficacy of the thymidine kinase suicide gene was confirmed using a live/dead assay. The epithelium was mechanically removed from athymic nude mice and remaining cells were treated with mitomycin C to prevent re-epithelialization. Telomerized corneal epithelial cells were seeded onto the denuded cornea and allowed to adhere for 4 and 24 hours. Cellular attachment was assessed using a fluorescent cell tracker. Stratification and differentiation were assessed after 7 days using phalloidin and a mouse monoclonal antibody to K3. RESULTS Telomerized corneal epithelial cells were visualized across the denuded stromal surface at 4 and 24 hours, with multi-layering evident at the latter time point. No epithelium was present in the non-treated eye. After 7 days post-transplantation cells stratified into a multilayered epithelium, with positive K3 expression in basal and suprabasal cells. Treatment with ganciclovir induced significant loss of viability in vitro. CONCLUSIONS The findings in this pilot study demonstrate that telomerized corneal epithelial cells possess the capacity to reconstitute a stratified corneal epithelium in vivo. The introduction of thymidine kinase allowed for the successful induction of cell death in proliferating cells in vitro. Collectively, these data suggest that a telomerase-immortalized corneal epithelial cell line transduced with thymidine kinase represents a potential model for studying differentiation and epithelial-niche interactions in vivo with potential applications in tissue engineering.
Collapse
Affiliation(s)
- Danielle M Robertson
- Department of Ophthalmology, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9057, USA.
| | | | | | | | | |
Collapse
|
13
|
Abstract
Secondary, so-called costimulatory, signals are critically required for the process of T cell activation. Since landmark studies defined that T cells receiving a T cell receptor signal without a costimulatory signal, are tolerized in vitro, the investigation of T cell costimulation has attracted intense interest. Early studies demonstrated that interrupting T cell costimulation allows attenuation of the alloresponse, which is particularly difficult to modulate due to the clone size of alloreactive T cells. The understanding of costimulation has since evolved substantially and now encompasses not only positive signals involved in T cell activation but also negative signals inhibiting T cell activation and promoting T cell tolerance. Costimulation blockade has been used effectively for the induction of tolerance in rodent models of transplantation, but turned out to be less potent in large animals and humans. In this overview we will discuss the evolution of the concept of T cell costimulation, the potential of 'classical' and newly identified costimulation pathways as therapeutic targets for organ transplantation as well as progress towards clinical application of the first costimulation blocking compound.
Collapse
Affiliation(s)
- Nina Pilat
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Austria
| | - Mohamed H. Sayegh
- Brigham and Women's Hospital & Children's Hospital Boston, Harvard Medical School, Boston, USA
| | - Thomas Wekerle
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Austria
| |
Collapse
|
14
|
Vieyra M, Leisman S, Raedler H, Kwan WH, Yang M, Strainic MG, Medof ME, Heeger PS. Complement regulates CD4 T-cell help to CD8 T cells required for murine allograft rejection. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:766-74. [PMID: 21704012 DOI: 10.1016/j.ajpath.2011.04.038] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/06/2011] [Accepted: 04/12/2011] [Indexed: 12/19/2022]
Abstract
Although induction of CD8 T-cell responses to transplants requires CD4-cell help, how this help is transmitted remains incompletely characterized. In vitro, cognate interactions between CD4 T cells and dendritic cells (DCs) induce C3a and C5a production. CD8(+) T cells lacking C3a receptor (C3aR) and C5a receptor (C5aR) proliferate weakly to allogeneic DCs despite CD4 help, indicating that CD4-cell help is mediated, in part, through DC-derived C3a/C5a acting on CD8(+) T cell-expressed C3aR/C5aR. In support of this concept, augmenting DC C5a/C3a production bypasses the requirement for CD4- and CD40-dependent help to wild-type CD8(+) T cells. CD4-deficient recipients of allogeneic heart transplants prime weak CD8 responses and do not acutely reject their grafts. In contrast, CD4-deficient chimeric mice possessing decay accelerating factor deficient (Daf1(-/-)) bone marrow, in which DC C3a/C5a production is potentiated, acutely reject transplants through a CD8 cell-dependent mechanism. Furthermore, hearts transplanted into CD40(-/-) mice prime weak CD8-cell responses and survive indefinitely, but hearts transplanted into Daf1(-/-)CD40(-/-) recipients undergo CD8 cell-dependent rejection. Together, the data indicate that heightened production and activation of immune cell-derived complement bypasses the need for CD40/CD154 interactions and implicate antigen-presenting cell-produced C5a and C3a as molecular bridges linking CD4 help to CD8(+) T cells.
Collapse
Affiliation(s)
- Mark Vieyra
- Renal Division, Department of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Lai YP, Lin CC, Liao WJ, Tang CY, Chen SC. CD4+ T cell-derived IL-2 signals during early priming advances primary CD8+ T cell responses. PLoS One 2009; 4:e7766. [PMID: 19901991 PMCID: PMC2770320 DOI: 10.1371/journal.pone.0007766] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 09/30/2009] [Indexed: 01/22/2023] Open
Abstract
Stimulating naïve CD8+ T cells with specific antigens and costimulatory signals is insufficient to induce optimal clonal expansion and effector functions. In this study, we show that the activation and differentiation of CD8+ T cells require IL-2 provided by activated CD4+ T cells at the initial priming stage within 0–2.5 hours after stimulation. This critical IL-2 signal from CD4+ cells is mediated through the IL-2Rβγ of CD8+ cells, which is independent of IL-2Rα. The activation of IL-2 signaling advances the restriction point of the cell cycle, and thereby expedites the entry of antigen-stimulated CD8+ T-cell into the S phase. Besides promoting cell proliferation, IL-2 stimulation increases the amount of IFNγ and granzyme B produced by CD8+ T cells. Furthermore, IL-2 at priming enhances the ability of P14 effector cells generated by antigen activation to eradicate B16.gp33 tumors in vivo. Therefore, our studies demonstrate that a full CD8+ T-cell response is elicited by a critical temporal function of IL-2 released from CD4+ T cells, providing mechanistic insights into the regulation of CD8+ T cell activation and differentiation.
Collapse
Affiliation(s)
- Yo-Ping Lai
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Ching Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Jung Liao
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Yung Tang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Ching Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
16
|
|
17
|
Mizuki N, Inoko H, Ohno S. Role of HLA and T lymphocytes in the immune response. Ocul Immunol Inflamm 2009; 2:57-91. [DOI: 10.3109/09273949409057064] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
18
|
Analysis of parathyroid graft rejection suggests alloantigen-specific production of nitric oxide by iNOS-positive intragraft macrophages. Transpl Immunol 2009; 21:183-91. [PMID: 19409993 DOI: 10.1016/j.trim.2009.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 04/17/2009] [Accepted: 04/24/2009] [Indexed: 01/18/2023]
Abstract
BACKGROUND During acute rejection of organ or tissue allografts T cells and macrophages are dominant infiltrating cells. CD4-positive T cells are important for the induction of allograft rejection and macrophages are important effector cells mediating cytotoxicity via production of nitric oxide (NO) by the inducible NO-synthase (iNOS). In the present study we analysed whether the destruction of primarily nonvascularised parathyroid allografts is also mediated by iNOS-positive macrophages. METHODS Hypocalcaemic Lewis rats received parathyroid isografts (from Lewis donors) and allografts (from Wistar Furth donors), respectively, under the kidney capsule. Levels of serum calcium above 2 mmol/L correlated with normal parathyroid function and below 2 mmol/L with parathyroid rejection. Accelerated parathyroid allograft rejection was induced by immunisation of Lewis recipients with the allogeneic peptide P1. RESULTS Determination of serum calcium levels is a useful parameter to control parathyroid graft function, and therefore to determine allograft rejection. Macrophages positive for both major histocompatibility complex (MHC) class II molecules and costimulatory molecules accumulated in iso- and allografts, but iNOS-positive macrophages were only detectable in allografts in the presence of activated CD4-positive T cells. These results confirm a cooperation between activated T cells and intragraft macrophages to induce macrophage iNOS expression. Recipients immunised with the allogeneic peptide P1 demonstrated accelerated rejection of allografts (mean+/-SD: 9.2+/-0.9 days) in contrast to nonimmunised animals (mean+/-SD: 15.8+/-1.8 days). Allografts of P1-immunised animals were infiltrated faster by activated CD4-positve T cells and, in addition, the infiltrates of iNOS-positive macrophages were stronger than those in allografts of nonimmunised animals. CONCLUSIONS Intragraft iNOS-positive macrophages seem to be able to produce cytotoxic NO involved in the killing of allogeneic cells during the alloimmune response against primarily nonvascularised parathyroid organ grafts. Infiltrates of iNOS-negative macrophages found in parathyroid isografts were caused by antigen-independent inflammation triggered by surgically induced injury. The absence of activated T cells in isografts and their presence in allografts underlines their importance in inducing macrophage iNOS expression.
Collapse
|
19
|
Hall BM, Tran G, Hodgkinson SJ. Alloantigen specific T regulatory cells in transplant tolerance. Int Immunopharmacol 2009; 9:570-4. [PMID: 19539571 DOI: 10.1016/j.intimp.2009.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 01/22/2009] [Indexed: 01/08/2023]
Abstract
CD4(+)CD25(+)Foxp3(+)T cells are regulatory/suppressor cells (Treg) that include non-antigen(Ag)-specific as well as Ag-specific Tregs. How non-Ag-specific naïve CD4(+)CD25(+)Treg develop into specific Tregs is unknown. We have studied DA rats tolerant to fully allogeneic PVG cardiac grafts that survived with out immunosuppression for over 100 days and identified the cellular basis of alloantigen specific tolerance. Key observations from our studies will be reviewed including how CD4(+)CD25(+)Tregs were first identified and the cytokine dependence of CD4(+)T cells that transfer alloantigen specific transplant tolerance which died in culture unless stimulated with both cytokine rich ConA supernatant and specific donor alloantigen. Both the tolerant CD4(+)CD25(+) and CD4(+)CD25(-) T cell populations are required to transfer tolerance, yet alone the CD4(+)CD25(-) T cell effect rejection. Tolerance transfer occurs with a low ratio of CD4(+)CD25(+)T cells (<1:10), whereas to induce tolerance with naive CD4(+)CD25(+)T cells requires both a ratio of >1:1 and is not alloantigen specific. Recent findings on how naïve CD4(+)CD25(+)T cells developed into two separated pathways of alloantigen specific Tregs, by culturing them with alloAg with either IL-2 or IL-4 and donor alloantigen are described. IL-2 enhances IFN-gammaR and IL-5 mRNA while IL-4 induced a reciprocal profile with de novo IL-5Ralpha and increased IFN-gamma mRNA expression. Both IL-2 and IL-4 alloactivated CD4(+)CD25(+)Tregs within 3-4 days of culture can induce alloantigen specific tolerance at ratios of 1:10. Long term, CD4(+)CD25(+)T cells from tolerant hosts given IL-2 cultured cells have increased IL-5 and IFN-gammaR mRNA; whereas hosts given IL-4 cultured cells had enhanced IL-5Ralpha mRNA expression and IL-5 enhanced their proliferation to donor but not third party alloAg. These findings suggest that Th1 and Th2 responses activate two pathways of alloantigen specific Tregs that can mediate transplant tolerance but are dependent upon cytokines produced by ongoing Th1 and/or Th2 immune responses.
Collapse
Affiliation(s)
- Bruce M Hall
- Department of Medicine, The University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia.
| | | | | |
Collapse
|
20
|
Richards DM, Zhang N, Dalheimer SL, Mueller DL. Allopeptide-specific CD4(+) T cells facilitate the differentiation of directly alloreactive graft-infiltrating CD8(+) T Cells. Am J Transplant 2007; 7:2269-78. [PMID: 17845562 DOI: 10.1111/j.1600-6143.2007.01934.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
To investigate the mechanism of CD4(+) T-cell help during the activation and differentiation of directly alloreactive CD8(+) T cells, we examined the development of obliterative airways disease (OAD) following transplantation of airways into fully mismatched recipient mice deficient in CD4(+) T cells. BALB/c trachea allografts became fibrosed significantly less frequently in B6 CD4(-/-) recipients as compared to wildtype controls. Furthermore, class I-directed cytotoxicity failed to develop in the absence of CD4(+) T cells. The infiltration of graft tissue by primed L(d)-specific directly alloreactive 2C CD8(+) T cells was not found to depend on the presence of CD4(+) T cells. Nevertheless, graft-infiltrating 2C CD8(+) T cells failed to express CD69 and granzyme B when CD4(+) T-cell help was unavailable. Importantly, reconstitution of B6 CD4(-/-) recipient mice with graft peptide-specific TCR-Tg CD4(+) T cells (OT-II or TEa) capable of recognizing antigen only on recipient APC allowed for full expression of CD69 and granzyme B by the directly alloreactive CD8(+) T cells and restored the capacity of recipients to reject their allografts. These results demonstrate that indirectly alloreactive CD4(+) T cells ensure the optimal activation and differentiation of graft-infiltrating directly alloreactive CD8(+) T cells independent of donor APC recognition.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- CD4 Antigens/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Cell Differentiation/immunology
- Graft Rejection/immunology
- Lectins, C-Type
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- T-Lymphocytes, Helper-Inducer/immunology
- Transplantation, Homologous/immunology
- Transplantation, Homologous/pathology
Collapse
Affiliation(s)
- D M Richards
- Department of Medicine, and Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | | | | | | |
Collapse
|
21
|
Ksander BR, Streilein JW. Recovery of activated cytotoxic T cells from minor H incompatible tumor graft rejection sites. 1989. Ocul Immunol Inflamm 2007; 15:205-13. [PMID: 17613835 DOI: 10.1080/09273940701404491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
22
|
Castellino F, Germain RN. Chemokine-guided CD4+ T cell help enhances generation of IL-6RalphahighIL-7Ralpha high prememory CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:778-87. [PMID: 17202339 DOI: 10.4049/jimmunol.178.2.778] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD4(+) T cells promote effective CD8(+) T cell-mediated immunity, but the timing and mechanistic details of such help remain controversial. Furthermore, the extent to which innate stimuli act independently of help in enhancing CD8(+) T cell responses is also unresolved. Using a noninfectious vaccine model in immunocompetent mice, we show that even in the presence of innate stimuli, CD4(+) T cell help early after priming is required for generating an optimal pool of functional memory CD8(+) T cells. CD4(+) T cell help increased the size of a previously unreported population of IL-6Ralpha(high)IL-7Ralpha(high) prememory CD8(+) T cells shortly after priming that showed a survival advantage in vivo and contributed to the majority of functional memory CD8(+) T cells after the contraction phase. In accord with our recent demonstration of chemokine-guided recruitment of naive CD8(+) T cells to sites of CD4(+) T cell-dendritic cell interactions, the generation of IL-6Ralpha(high)IL-7Ralpha(high) prememory as well as functional memory CD8(+) T cells depended on the early postvaccination action of the inflammatory chemokines CCL3 and CCL4. Together, these findings support a model of CD8(+) T cell memory cell differentiation involving the delivery of key signals early in the priming process based on chemokine-guided attraction of naive CD8(+) T cells to sites of Ag-driven interactions between TLR-activated dendritic cells and CD4(+) T cells. They also reveal that elevated IL-6Ralpha expression by a subset of CD8(+) T cells represents an early imprint of CD4(+) T cell helper function that actively contributes to the survival of activated CD8(+) T cells.
Collapse
Affiliation(s)
- Flora Castellino
- Lymphocyte Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | | |
Collapse
|
23
|
Abstract
Concepts of cell-cell interactions in adaptive immunity have alternated between the simple and the complex. The notion that one population of small, circulating lymphocytes is responsible for adaptive immunity was sequentially supplanted by the concept of separate T and B lymphocyte populations that cooperate to produce IgG antibody responses, by a three-cell model in which a myeloid APC initiates these cooperative lymphoid responses, by the recognition of T cell subsets, and by the idea that CD8+ T cell subset responses to graft antigens depend on CD4+ T cell subset activity. Simplicity was reintroduced with the revelation that CD8+ T cells can act independently of CD4+ T cells against acute viral infections. The pendulum has swung again toward complexity with recognition of the distinct and conjoint contributions of innate stimuli, APCs, NK and NKT cells, Tregs, and CD4+ helper T cells to CD8+ T cell behavior during acute and chronic infections or as memory cells. The renewed appreciation that multiple, sometimes rare cell types must communicate during cell-mediated immune responses has led to questions about how such interactions are orchestrated within organized lymphoid tissues. We review recent advances in deciphering the specific contribution of CD4+ T cells to physiologically useful CD8+ T cell responses, the signals involved in producing acute effectors versus long-lived memory cells, and the mechanisms underlying the cell-cell associations involved in delivery of such signals. We propose a model based on these new findings that may serve as a general paradigm for cellular interactions that occur in an inflamed lymph node during the initiation of immune responses.
Collapse
Affiliation(s)
- Flora Castellino
- Lymphocyte Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
24
|
Chen BG, Liu Z, Wu Y. Specific tolerance induction of allo-K(b)-skin grafts by FK506 in the CD8-depleted H-2(k) recipients required low amounts of K(b)-antigen. Transpl Immunol 2005; 15:9-16. [PMID: 16223668 DOI: 10.1016/j.trim.2005.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Accepted: 02/15/2005] [Indexed: 11/29/2022]
Abstract
MHC class I allo-grafts can be directly rejected by recipient CD8 T cells and be indirectly rejected by recipient CD4 T cells. Although the experimental results using the bm mutant and C57BL/6 mice indicated that CD4-mediated rejection of class I-disparate grafts is a relatively weak process and is expected to be more sensitive to additional exogenous immunosuppression, it is unclear that whether this mechanism can be used for inducing a specific tolerance of class I disparate grafts. In this study, we hypothesize that a short course of FK506 may induce a specific tolerance of class I-disparate skin grafts in the CD8-depleted recipients. K(b)-transgenic C3H mice, Tg.H-2 K(b)-1 and Tg.H-2 K(b)-2 mice that express high copies and low copies of K(b)-antigen respectively were used as donors. Wild type C3H mice (H-2(k)) in which either CD4 or CD8 T cells were depleted by administration of anti-CD4 or CD8 monoclonal antibody (mAb) were used as recipients. Results showed that FK506 promoted longer survival of allo-K(b) skin grafts in CD8-depleted C3H mice than in CD4-depleted C3H mice. Graft survival from Tg.H-2 K(b)-2 mice was significantly longer than Tg.H-2 K(b)-1 mice. A short course of FK506 induced long-term survival of skin grafts from Tg.H-2K(b)-2 mice, but not from Tg.H-2K(b)-1 mice in CD8-depleted C3H recipients, even after FK506 was stopped. These mice also accepted grafts of Tg.H-2K(b)-1 mice when challenged with skin grafts from Tg.H-2K(b)-1 mice, but promptly rejected third party skin grafts from BALB/c (H-2(d)) mice. T cells from K(b)-tolerant C3H mice did not respond to allo-K(b)-antigen in in vitro assays of mixed lymphocyte culture and cell-mediated cytotoxicity. In conclusion we found that a short course of FK506 treatment and low amounts of K(b)-antigen induced a K(b)-specific tolerance in the CD8-depleted recipients, and this tolerance maintained even after withdrawing the anti-CD8 mAb treatment.
Collapse
Affiliation(s)
- Bing-Guan Chen
- Medical Research Center, Tongji University Affiliated Shanghai East Hospital, 150 Jimo Road, Shanghai 200120, PR China.
| | | | | |
Collapse
|
25
|
Estcourt MJ, McMichael AJ, Hanke T. Altered primary CD8+ T cell response to a modified virus Ankara(MVA)-vectored vaccine in the absence of CD4+ T cell help. Eur J Immunol 2005; 35:3460-7. [PMID: 16245361 DOI: 10.1002/eji.200526284] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
T cell receptor-transgenic F5 mice were used to assess primary CD8+ T cell responses to a modified virus Ankara (MVA)-vectored vaccine in the absence of CD4+ T cell help. Naive, CD8-enriched, CFSE-labelled F5 cells were transferred into normal or CD4+ cell-depleted mice and the mice were vaccinated with MVA.HIVA-NP. At different time points during the primary response, F5 cells were re-isolated and analysed on divisional basis for a number of parameters. We demonstrated that the primary CD8+ T cell response in the absence of CD4+ T cell help differed from that in normal CD4+ cell-undepleted mice. While in the absence of CD4+ T cell help, the initial migratory progress from the local response to a systemic one was not grossly affected, the proportion of dying F5 cells during the expansion phase was markedly increased and resulted in an overall smaller expansion and significantly decreased frequency of CD8+ T cell memory after contraction. T cells primed without help displayed accelerated proliferation and activation, while expression of interferon-gamma remained similar. These phenomena were observed in the lymph nodes draining the MVA.HIVA-NP immunization site and were similar, but delayed by 2-3 days in spleen and non-draining lymph nodes.
Collapse
Affiliation(s)
- Marie J Estcourt
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The John Radcliffe, Oxford, UK
| | | | | |
Collapse
|
26
|
Brehm MA, Daniels KA, Ortaldo JR, Welsh RM. Rapid conversion of effector mechanisms from NK to T cells during virus-induced lysis of allogeneic implants in vivo. THE JOURNAL OF IMMUNOLOGY 2005; 174:6663-71. [PMID: 15905505 DOI: 10.4049/jimmunol.174.11.6663] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Viral infections can strongly stimulate both NK cell and allospecific CD8 T cell responses, and these same effector cells can lyse allogeneic cell lines in vitro. However, the impact of viral infections on the effector systems mediating rejection of allogeneic tissues in vivo has not been fully explored. Using in vivo cytotoxicity assays, we evaluated the effector systems mediating the rejection of CFSE-labeled allogeneic splenocytes after an infection of C57BL/6 (B6) mice with lymphocytic choriomeningitis virus. Naive B6 mice predominantly used a NK cell-effector mechanism to reject allogeneic splenocytes because they rejected BALB/C (H2(d)) splenocytes but not CBA (H2(k)) splenocytes, and the rejection was prevented by immunodepletion of NK1.1(+) or Ly49D(+) NK cells. This rapid and efficient in vivo cytotoxicity assay recapitulated the specificity of NK cell-mediated rejection seen in longer duration in vivo assays. However, as early as 1 day after infection with lymphocytic choriomeningitis virus, a CD8 T cell-dependent mechanism participated in the rejection process and a broader range of tissue haplotypes (e.g., H2(k)) was susceptible. The CD8 T cell-mediated in vivo rejection process was vigorous at a time postinfection (day 3) when NK cell effector functions are peaking, indicating that the effector systems used in vivo differed from those observed with in vitro assays measuring the killing of allogeneic cells. This rapid generation of allospecific CTL activity during a viral infection preceded the peak of viral epitope-specific T cell responses, as detected by in vivo or in vitro cytotoxicity assays.
Collapse
Affiliation(s)
- Michael A Brehm
- Department of Pathology, University of Massachusetts Medical School, Worcester, 01655, USA
| | | | | | | |
Collapse
|
27
|
Xiang J, Huang H, Liu Y. A New Dynamic Model of CD8+T Effector Cell Responses via CD4+T Helper-Antigen-Presenting Cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:7497-505. [PMID: 15944248 DOI: 10.4049/jimmunol.174.12.7497] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A long-standing paradox in cellular immunology has been the conditional requirement for CD4(+) Th cells in priming of CD8(+) CTL responses. We propose a new dynamic model of CD4(+) Th cells in priming of Th-dependent CD8(+) CTL responses. We demonstrate that OT II CD4(+) T cells activated by OVA-pulsed dendritic cells (DC(OVA)) are Th1 phenotype. They acquire the immune synapse-composed MHC II/OVAII peptide complexes and costimulatory molecules (CD54 and CD80) as well as the bystander MHC class I/OVAI peptide complexes from the DC(OVA) by DC(OVA) stimulation and thus also the potential to act themselves as APCs. These CD4(+) Th-APCs stimulate naive OT I CD8(+) T cell proliferation through signal 1 (MHC I/OVAI/TCR) and signal 2 (e.g., CD54/LFA-1 and CD80/CD28) interactions and IL-2 help. In vivo, they stimulate CD8(+) T cell proliferation and differentiation into CTLs and induce effective OVA-specific antitumor immunity. Taken together, this study demonstrates that CD4(+) Th cells carrying acquired DC Ag-presenting machinery can, by themselves, efficiently stimulate CTL responses. These results have substantial implications for research in antitumor and other aspects of immunity.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Bystander Effect/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line, Tumor
- Cell Proliferation
- Cytotoxicity, Immunologic/genetics
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class II/metabolism
- Hybridomas
- Intercellular Adhesion Molecule-1/metabolism
- Lymphocyte Activation/immunology
- Melanoma, Experimental
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Models, Immunological
- Molecular Sequence Data
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/prevention & control
- Ovalbumin/immunology
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Jim Xiang
- Research Unit, Saskatchewan Cancer Agency, Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | | | | |
Collapse
|
28
|
Youssef AR, Otley C, Mathieson PW, Smith RM. Role of CD4+ and CD8+ T cells in murine skin and heart allograft rejection across different antigenic desparities. Transpl Immunol 2005; 13:297-304. [PMID: 15589743 DOI: 10.1016/j.trim.2004.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2004] [Accepted: 10/25/2004] [Indexed: 11/17/2022]
Abstract
The factors that influence the relative contribution of the T cell subsets to allograft rejection remain unclear. We compared skin and heart rejection in CD4 Knockout (KO), and CD8 KO mice across full-, minor-, and class II histocompatibility antigen (HA) mismatches. Skin allografts were rejected by either CD4+ or CD8+ T cells alone at any degree of antigenic mismatch. However, either the absence of CD4+ cells or a lesser degree of HA mismatch resulted in prolongation of graft survival. In contrast, fully allogeneic heart grafts were accepted in CD4 KO recipients, and minor HA mismatched heart grafts were accepted by both CD4 KO and CD8 KO mice. Thus, the T cell subsets required for allograft rejection are determined by the immunogenicity of the tissue transplanted. In the absence of CD8+ T cells, perforin and Fas ligand (FasL) but not granzyme B mRNA were detected in rejecting grafts. Thus, granzyme B is a CD8+ cytotoxic T lymphocyte (CTL)-specific effector molecule.
Collapse
Affiliation(s)
- Abdel-Rahman Youssef
- Academic Renal Unit, Southmead Hospital, University of Bristol, Bristol BS10 5NB, UK
| | | | | | | |
Collapse
|
29
|
Cortesini NSF, Colovai AI, Manavalan JS, Galluzzo S, Naiyer AJ, Liu J, Vlad G, Kim-Schulze S, Scotto L, Fan J, Cortesini R. Role of regulatory and suppressor T-cells in the induction of ILT3+ ILT4+ tolerogenic endothelial cells in organ allografts. Transpl Immunol 2005; 13:73-82. [PMID: 15380537 DOI: 10.1016/j.trim.2004.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Accepted: 06/03/2004] [Indexed: 11/16/2022]
|
30
|
Melchionda F, McKirdy MK, Medeiros F, Fry TJ, Mackall CL. Escape from immune surveillance does not result in tolerance to tumor-associated antigens. J Immunother 2005; 27:329-38. [PMID: 15314541 DOI: 10.1097/00002371-200409000-00001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Despite expression of tumor-associated or tumor-specific antigens by most tumors, evasion of protective T-cell immunity is the rule rather than the exception. Understanding whether tumor immune escape primarily represents T-cell neglect, anergy/tolerance, or quantitative limits of an existent immune response is central to developing new strategies to enhance antitumor immunity. The authors studied the immune response to MB49, a tumor that naturally expresses HY. Immune surveillance was effective following low-dose tumor inocula, since normal female mice showed a diminished incidence and slower growth rate of MB49 compared with T-cell-depleted female mice and male mice. Following high-dose tumor inoculation, females developed large, progressive tumors but continued to demonstrate immune responses to class I and class II restricted HY epitopes. The HY reactive T cells remained capable of executing HY immune responses since T cells adoptively transferred from MB49-bearing animals mediated accelerated HY skin graft rejection compared with those taken from naive mice. Thus, MB49 does not induce immune tolerance to HY but rather escapes immune surveillance largely due to quantitative limits of the immune response. Treatment of tumor-bearing animals with rhIL7 significantly increased the number of T cells responding to HY but did not alter tumor growth rate. These results demonstrate that escape from immune surveillance does not necessarily imply immune tolerance to tumor antigens and that immunotherapy need not overcome tumor-induced tolerance per se, and suggest that substantial opportunities remain in tumor-bearing hosts to amplify weak but persistent antitumor immune responses.
Collapse
Affiliation(s)
- Fraia Melchionda
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|
31
|
Abstract
Antigens, provided by the allograft, trigger the activation and proliferation of allospecific T cells. As a consequence of this response, effector elements are generated that mediate graft injury and are responsible for the clinical manifestations of allograft rejection. Donor-specific CD8+ cytotoxic T lymphocytes play a major role in this process. Likewise, CD4+ T cells mediate delayed-type hypersensitivity responses via the production of soluble mediators that function to further activate and guide immune cells to the site of injury. In addition, these mediators may directly alter graft function by modulating vascular tone and permeability or by promoting platelet aggregation. Allospecific CD4+ T cells also promote B-cell maturation and differentiation into antibody-secreting plasma cells via CD40-CD40 ligand interactions. Alloantibodies that are produced by these B cells exert most of their detrimental effects on the graft by activating the complement cascade. Alternatively, antibodies can bind Fc receptors on natural killer cells or macrophages and cause target cell lysis via antibody-dependent cell-mediated cytotoxicity. In this review, we discuss these major effector pathways, focusing on their role in the pathogenesis of allograft rejection.
Collapse
Affiliation(s)
- Paulo N Rocha
- Duke University and Durham VA Medical Centers, Durham, NC 27705, USA
| | | | | | | |
Collapse
|
32
|
Wang JCE, Livingstone AM. Cutting edge: CD4+ T cell help can be essential for primary CD8+ T cell responses in vivo. THE JOURNAL OF IMMUNOLOGY 2004; 171:6339-43. [PMID: 14662830 DOI: 10.4049/jimmunol.171.12.6339] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent studies have shown that CD4(+) T cell help is required for the generation of memory CD8(+) T cells that can proliferate and differentiate into effector cells on Ag restimulation. The importance of help for primary CD8(+) T cell responses remains controversial. It has been suggested that help is not required for the initial proliferation and differentiation of CD8(+) T cells in vivo and that classical models of helper-dependent responses describe impaired secondary responses to Ag in vitro. We have measured primary CD8(+) T cell responses to peptide-pulsed dendritic cells in mice by cytokine ELISPOT and tetramer staining. No responses were detected in the absence of help, either when normal dendritic cells were injected into MHC II-deficient mice or when MHC II-deficient dendritic cells were injected into normal mice. Thus, the primary in vivo CD8(+) T cell response depends absolutely on help from CD4(+) T cells in our experimental system.
Collapse
Affiliation(s)
- Jyh-Chiang E Wang
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, and Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | |
Collapse
|
33
|
Uenaka A, Nakayama E. Murine leukemia RL male 1 and sarcoma Meth A antigens recognized by cytotoxic T lymphocytes (CTL). Cancer Sci 2003; 94:931-6. [PMID: 14611667 PMCID: PMC11160269 DOI: 10.1111/j.1349-7006.2003.tb01380.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2003] [Revised: 09/02/2003] [Accepted: 09/05/2003] [Indexed: 11/29/2022] Open
Abstract
Peptide elution and expression cloning methods have been used to identify T cell-recognized antigens for which no molecular information is available. We identified a unique tumor antigen peptide pRL1a, IPGLPLSL that is recognized by CTL on BALB/c RL male 1 leukemia by peptide elution. The sequence of the peptide corresponded to the normally untranslated 5' region of akt. Cytotoxicity was generated in BALB/c spleen cells by in vivo and in vitro sensitization with pRL1a peptide in the form of multiple antigen peptide (MAP), but not the original form. pRL1a MAP immunization had a significant growth-inhibitory effect. pRL1a MAP was mostly internalized into the endosomal compartment of antigen-presenting cells, leaked to the cytosol, and degraded, and the pRL1a peptide produced was presented through the MHC class I pathway. In vivo depletion of CD4 T cells from tumor-inoculated BALB/c mice caused RL male 1 regression. Overexpression of the RLakt molecule seemed to induce CD4 immunoregulatory cells, which resulted in progressive RL male 1 growth in BALB/c mice. In vivo administration of anti-CD25 mAb (PC61) caused regression of RL male 1, suggesting that CD4(+) CD25(+) immunoregulatory cells were involved in the tumor growth. Recently, we improved the sensitivity and the efficacy of T cell antigen cloning from cDNA expression libraries by using large- and small-scale ELISPOT assays. Using the IFN-gamma ELISPOT method, we obtained a cDNA clone S35 of 937 bp recognized by AT-1 CTL on BALB/c Meth A sarcoma. S35 was a part of the retinoic acid-regulated nuclear matrix-associated protein (ramp). AT-1 CTL recognized the peptide LGAEAIFRL, which was derived from a newly created open reading frame due to the exon 14 extension.
Collapse
Affiliation(s)
- Akiko Uenaka
- Department of Immunology, Okayama University Graduate School of Medicine and Dentistry, Okayama 700-8558.
| | | |
Collapse
|
34
|
Csencsits KL, Bishop DK. Contrasting alloreactive CD4+ and CD8+ T cells: there's more to it than MHC restriction. Am J Transplant 2003; 3:107-15. [PMID: 12603205 DOI: 10.1034/j.1600-6143.2003.00036.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Surface expression of CD4 or CD8 is commonly used to identify T-cell subsets that recognize antigen presented by class II MHC or class I MHC, respectively. This holds true for T cells that respond to allogeneic MHC molecules that are directly recognized as foreign, as well as peptides from allogeneic MHC molecules that are indirectly presented by self MHC molecules. CD4 or CD8 expression was initially believed to define cytokine secreting helper T cells or cytotoxic cells, respectively. However, this association of phenotype and function is not absolute, in that CD4+ cells may possess lytic activity and CD8+ cells secrete cytokines, notably IFNgamma. Recently, additional fundamental differences in the immunobiology of these T-cell subsets have been identified. These include differences in costimulatory requirements, cytokine responsiveness, cytokine production, cell survival, and the maintenance of memory. This review will survey these differences, emphasizing alloreactive T-cell responses as well as relevant observations that have been made in other systems.
Collapse
Affiliation(s)
- Keri L Csencsits
- Depajtment of Surgery University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
35
|
Illigens BM, Yamada A, Fedoseyeva EV, Anosova N, Boisgerault F, Valujskikh A, Heeger PS, Sayegh MH, Boehm B, Benichou G. The relative contribution of direct and indirect antigen recognition pathways to the alloresponse and graft rejection depends upon the nature of the transplant. Hum Immunol 2002; 63:912-25. [PMID: 12368044 DOI: 10.1016/s0198-8859(02)00449-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, we measured direct and indirect T-cell alloresponses mediated by CD4(+) and CD8(+) T cells in three mouse transplantation models: skin, cornea, and retina. We show that the contribution of direct and indirect antigen recognition pathways to the alloresponse to fully allogeneic grafts varies depending upon the nature of the tissue/organ transplanted. The implications of this finding for understanding the cellular mechanisms by which rejection is mediated in different transplant models are discussed.
Collapse
Affiliation(s)
- Ben M Illigens
- Cellular and Molecular Immunology Laboratory, Schepens Eye Research Institute and Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Surquin M, Le Moine A, Flamand V, Nagy N, Rombaut K, Demoor FX, Stordeur P, Salmon I, Guéry JC, Goldman M, Abramowicz D. Skin graft rejection elicited by beta 2-microglobulin as a minor transplantation antigen involves multiple effector pathways: role of Fas-Fas ligand interactions and Th2-dependent graft eosinophil infiltrates. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:500-6. [PMID: 12077281 DOI: 10.4049/jimmunol.169.1.500] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Beta(2)-microglobulin (beta(2)m)-derived peptides are minor transplantation Ags in mice as beta(2)m-positive skin grafts (beta(2)m(+/+)) are rejected by genetically beta(2)m-deficient recipient mice (beta(2)m(-/-)). We studied the effector pathways responsible for the rejection induced by beta(2)-microglobulin-derived minor transplantation Ags. The rejection of beta(2)m(+/+) skin grafts by naive beta(2)m(-/-) mice was dependent on both CD4 and CD8 T cells as shown by administration of depleting mAbs. Experiments performed with beta(2)m(-/-)CD8(-/-) double knockout mice grafted with a beta(2)m(+/+) MHC class I-deficient skin showed that sensitized CD4 T cells directed at beta(2)m peptides-MHC class II complexes are sufficient to trigger rapid rejection. Rejection of beta(2)m(+/+) grafts was associated with the production of IL-5 in vitro, the expression of IL-4 and IL-5 mRNAs in the grafted tissue, and the presence within rejected grafts of a considerable eosinophil infiltrate. Blocking IL-4 and IL-5 in vivo and depleting eosinophils with an anti-CCR3 mAb prevented graft eosinophil infiltration and prolonged beta(2)m(+/+) skin graft survival. Lymphocytes from rejecting beta(2)m(-/-) mice also displayed an increased production of IFN-gamma after culture with beta(2)m(+/+) minor alloantigens. In vivo neutralization of IFN-gamma inhibited skin graft rejection. Finally, beta(2)m(+/+) skin grafts harvested from B6(lpr/lpr) donor mice, which lack a functional Fas molecule, survived longer than wild-type beta(2)m(+/+) skin grafts, showing that Fas-Fas ligand interactions are involved in the rejection process. We conclude that IL-4- and IL-5-dependent eosinophilic rejection, IFN-gamma-dependent mechanisms, and Fas-Fas ligand interactions are effector pathways in the acute rejection of minor transplantation Ags.
Collapse
Affiliation(s)
- Murielle Surquin
- Laboratory of Experimental Immunology, Université Libre de Bruxelles, and Department of Nephrology, Hôpital Erasme, Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kreisel D, Krupnick AS, Gelman AE, Engels FH, Popma SH, Krasinskas AM, Balsara KR, Szeto WY, Turka LA, Rosengard BR. Non-hematopoietic allograft cells directly activate CD8+ T cells and trigger acute rejection: an alternative mechanism of allorecognition. Nat Med 2002; 8:233-9. [PMID: 11875493 DOI: 10.1038/nm0302-233] [Citation(s) in RCA: 184] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite evidence that human non-hematopoietic cells, such as vascular endothelium, can activate allogeneic T lymphocytes in vitro, the prevailing view has been that hematopoietic antigen-presenting cells are required to trigger alloimmune responses in vivo. Here we report that mouse non-hematopoietic cells activate alloreactive CD8+ T lymphocytes in vitro and in vivo. We also show that vascularized cardiac allografts are acutely rejected via CD8+ direct allorecognition even if the alloantigen is not presented by hematopoietic professional antigen-presenting cells. Because activation of alloreactive CD8+ T cells by donor-type non-hematopoietic cells can continue for the life of the allograft, these findings present a new clinically relevant mechanism of allorecognition and should be taken into consideration when developing strategies to prevent allograft vasculopathy or to induce tolerance.
Collapse
Affiliation(s)
- Daniel Kreisel
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Halamay KE, Kirkman RL, Sun L, Yamada A, Fragoso RC, Shimizu K, Mitchell RN, McKay DB. CD8 T cells are sufficient to mediate allorecognition and allograft rejection. Cell Immunol 2002; 216:6-14. [PMID: 12381345 DOI: 10.1016/s0008-8749(02)00530-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Different T cell subsets may play different roles in allorecognition and allograft rejection. It has been suggested that CD8 T cells can only initiate rejection with help from CD4 T cells. Since CD8 T cells may have different requirements for allorecognition and for costimulation, it is important to clarify the role of CD8 cells in rejection. We examined the role of CD8 cells in allorecognition using a TCR transgenic mouse transplantation model. In our study, CD8 cells were able to recognize alloantigens and reject allografts in the absence of help from CD4 T cells. Furthermore our study provides a model to study the mechanisms of CD8-mediated allograft rejection. It may be important in the future, to consider that CD8 T cells may need to be targeted independently of CD4 T cells in strategies used to prevent rejection and induce tolerance.
Collapse
Affiliation(s)
- Kate E Halamay
- Division of Pediatrics, Department of Pediatric Oncology, Dana Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Boisgérault F, Liu Y, Anosova N, Ehrlich E, Dana MR, Benichou G. Role of CD4+ and CD8+ T cells in allorecognition: lessons from corneal transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:1891-9. [PMID: 11489968 DOI: 10.4049/jimmunol.167.4.1891] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Corneal transplantation represents an interesting model to investigate the contribution of direct vs indirect Ag recognition pathways to the alloresponse. Corneal allografts are naturally devoid of MHC class II+ APCs. In addition, minor Ag-mismatched corneal grafts are more readily rejected than their MHC-mismatched counterparts. Accordingly, it has been hypothesized that these transplants do not trigger direct T cell alloresponse, but that donor Ags are presented by host APCs, i.e., in an indirect fashion. Here, we have determined the Ag specificity, frequency, and phenotype of T cells activated through direct and indirect pathways in BALB/c mice transplanted orthotopically with fully allogeneic C57BL/6 corneas. In this combination, only 60% of the corneas are rejected, while the remainder enjoy indefinite graft survival. In rejecting mice the T cell response was mediated by two T cell subsets: 1) CD4+ T cells that recognize alloantigens exclusively through indirect pathway and secrete IL-2, and 2) IFN-gamma-producing CD8+ T cells recognizing donor MHC in a direct fashion. Surprisingly, CD8+ T cells activated directly were not required for graft rejection. In nonrejecting mice, no T cell responses were detected. Strikingly, peripheral sensitization to allogeneic MHC molecules in these mice induced acute rejection of corneal grafts. We conclude that only CD4+ T cells activated via indirect allorecognition have the ability to reject allogeneic corneal grafts. Although alloreactive CD8+ T cells are activated via the direct pathway, they are not fully competent and cannot contribute to the rejection unless they receive an additional signal provided by professional APCs in the periphery.
Collapse
Affiliation(s)
- F Boisgérault
- Cellular and Molecular Immunology Laboratory, Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | |
Collapse
|
40
|
Matsumiya G, Shirakura R, Miyagawa S, Izutani H, Nakata S, Shimazaki Y, Nakano S, Matsuda H. Role of CD4+ T cells in the rat to mouse cardiac xenotransplantation. Transpl Int 2001; 7 Suppl 1:S654-9. [PMID: 11271332 DOI: 10.1111/j.1432-2277.1994.tb01466.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
T cell subsets involved in rejection of xenografts were analyzed using a rat to mouse cardiac xenotransplant model. Proliferating response and interleulin-2 (IL-2) production in recipients' spleen cells were almost completely abrogated by elimination of L3T4+ T cells, but not by elimination of Lyt2.1+ T cells. Cytotoxic T lymphocyte (CTL) activities were mediated by both L3T4+ and Lyt2.1+ T cells with the help of IL-2-producing L3T4+ T cells. Administration of anti-L3T4 monoclonal antibody (mAb) into recipient mice resulted in a significant prolongation of graft survival (mean graft survival was 29.2 days). Moreover, anti-L3T4 mAb treatment plus thymectomy led to indefinite graft survival. Anti-rat endothelial cell (EC) antibody production in the grafted mice was remarkably suppressed by anti-L3T4 mAb treatment. In contrast, Lyt2.1 mAb treatment did not prolong the graft survival and did not suppress anti-EC antibody production. These results indicated the absolute requirement of L3T4+ T cells in the rejection of rat to mouse cardiac xenografts.
Collapse
Affiliation(s)
- G Matsumiya
- First Department of Surgery, Osaka University Medical School, Suita, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Anderson CC, Carroll JM, Gallucci S, Ridge JP, Cheever AW, Matzinger P. Testing time-, ignorance-, and danger-based models of tolerance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:3663-71. [PMID: 11238605 DOI: 10.4049/jimmunol.166.6.3663] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this study, we present data showing that tolerance to Ags in the periphery is not determined by the time at which the Ag appears, or by special properties of tissues in newborn mice or newly developing immune systems. We placed male grafts onto immunoincompetent female mice, allowed the grafts to heal for up to 5 mo, and then repopulated the recipients with fetal liver stem cells. We found that the newly arising T cells were neither tolerant nor ignorant of the grafts, but promptly rejected them, though they did not reject female grafts, nor show any signs of autoimmunity. We also found that the H-Y Ag was continuously cross-presented on host APCs, that this presentation was immunogenic, not tolerogenic, and that it depended on the continuous presence of the graft. In searching for the stimulus that might activate the host APCs, we analyzed mRNA expression with a highly sensitive real-time quantitative PCR assay. By using two different "housekeeping" molecules for comparison, we analyzed the message levels for several stress and/or inflammatory molecules in the healed grafts. We found that the long-healed grafts were not equivalent to "normal" skin because the healed grafts expressed lower levels of GAPDH. Altogether, these data suggest that acceptance vs rejection of peripheral tissues is not attributable to ignorance, timing-based tolerance, or special circulation properties of naive T cells in neonatal tissues. It is more likely attributable to an aspect of the context of Ag presentation that remains to be identified.
Collapse
Affiliation(s)
- C C Anderson
- Ghost Lab, Section on T cell Tolerance and Memory, Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Anderson CC, Matzinger P. Immunity or tolerance: opposite outcomes of microchimerism from skin grafts. Nat Med 2001; 7:80-7. [PMID: 11135620 DOI: 10.1038/83393] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Solid organ transplants contain small numbers of leukocytes that can migrate into the host and establish long-lasting microchimerism. Although such microchimerism is often associated with graft acceptance and tolerance, it has been difficult to demonstrate a true causal link. Using skin from mutant mice deficient for leukocyte subsets, we found that donor T-cell chimerism is a 'double-edged sword' that can result in very different outcomes depending on the host's immunological maturity and the antigenic disparities involved. In immunologically mature hosts, chimerism resulted in immunity and stronger graft rejection. In immature hosts, it resulted in tolerance to the chimeric T cells, but not to graft antigens not expressed by the chimeric cells. Clinical efforts aimed at augmenting chimerism to induce tolerance must take into account the maturation state of host T cells, the type of chimerism produced by each organ and the antigenic disparities involved, lest the result be increased rejection rather than tolerance.
Collapse
Affiliation(s)
- C C Anderson
- Ghost Lab, Section on T-cell Tolerance and Memory, Laboratory of Cellular and Molecular Immunology, NIAID/NIH Building 4 Room 111, 9000 Rockville Pike, Bethesda, Maryland 20892-0420, USA.
| | | |
Collapse
|
43
|
New Insights into Prevention of Donor-specific Corneal Graft Rejection. Cornea 2000. [DOI: 10.1097/00003226-200011003-00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
|
45
|
Borenstein SH, Graham J, Zhang XL, Chamberlain JW. CD8+ T cells are necessary for recognition of allelic, but not locus-mismatched or xeno-, HLA class I transplantation antigens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:2341-53. [PMID: 10946256 DOI: 10.4049/jimmunol.165.5.2341] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although HLA transgenic mice (HLA TgM) could provide a powerful approach to investigate human MHC-specific T cell responsiveness, the extent to which these molecules are recognized by the mouse immune system remains unclear. We established TgM expressing HLA class I alleles A2, B7, or B27 in their fully native form (HLAnat) or as hybrid molecules (HLAhyb) of the HLA alpha1/alpha2 domains linked to the H-2Kb alpha3, transmembrane, and cytoplasmic domains (i.e., to maintain possible species-specific interactions). Comparison of each as xeno- (i.e., by non-TgM) vs allo- (i.e., by TgM carrying an alternate HLA allele) transplantation Ags revealed the following: 1) Although HLAhyb molecules induced stronger xeno-CD8+ T cell responses in vitro, additional effector mechanisms must be active in vivo because HLAnat skin grafts were rejected faster by non-TgM; 2) gene knockout recipients showed that xenorejection of HLAnat and, unexpectedly, HLAhyb grafts doesn't depend on CD8+ or CD4+ T cells or B cells; 3) each HLAhyb strain developed tolerance to "self" but rejected allele- (-B27 vs -B7) and locus- (-B vs -A) mismatched grafts, the former requiring CD8+ T cells, the latter by CD8+ T cell-independent mechanisms. The finding that recognition of xeno-HLAhyb does not require CD8+ T cells while recognition of the identical molecule in a strictly allo context does, demonstrates an alpha1/alpha2 domain-dependent difference in effector mechanism(s). Furthermore, the CD8+ T cell-independence of locus-mismatched rejection suggests the degree of similarity between self and non-self alpha1/alpha2 determines the effector mechanism(s) activated. The HLA Tg model provides a unique approach to characterize these mechanisms and develop tolerance protocols in the context of human transplantation Ags.
Collapse
Affiliation(s)
- S H Borenstein
- Institute of Medical Science, University of Toronto, The Hospital for Sick Children, Toronto, Canada
| | | | | | | |
Collapse
|
46
|
Jiang H, Yang X, Soriano RN, Fujimura T, Krishnan K, Kobayashi M. Distinct patterns of cytokine gene suppression by the equivalent effective doses of cyclosporine and tacrolimus in rat heart allografts. Immunobiology 2000; 202:280-92. [PMID: 11045663 DOI: 10.1016/s0171-2985(00)80034-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In vitro studies of the mode of action of cyclosporine (CsA) and tacrolimus have indicated that both drugs produce immunosuppression by a quite similar cellular and molecular mechanism to block T cell receptor emanated transcriptional activation of interleukin(IL)-2 and other cytokine genes. Herein, we show that there are distinct patterns of cytokine gene expression in rat heart allografts under equivalent effective doses ("optimal dose") of CsA and tacrolimus. The optimal doses of CsA (10 mg/kg/day) and tacrolimus (3.2 mg/kg/day), which induce similar mean graft survival time (MST), were administered in LEW recipients with ACI heart grafts from day 0 after grafting until sacrifice. Heart grafts were harvested at days 3, 5, and 7. The expression of various cell surface markers, cytokines, and cytotoxic factors was determined by immunohistology and reverse transcriptase-polymerase chain reaction (RFT-PCR). Cell populations that stained positively in the heart tissues of allograft control increased through day 7 for CD4+ and CD8+ T lymphocytes, NKR-Pla+ natural killer (NK) cells, and ED2+ macrophages. CsA and tacrolimus have comparable activity to block these cell local infiltrations. The mRNA levels of the majority of the factors were dramatically up-regulated in the allografts over time, peaking at day 5. The optimal doses of CsA and tacrolimus had similar inhibitory effects on Th1 type cytokine IL-2 and interferon [INF]-gamma), inflammatory cytokine (IL-1beta and tumor necrosis factor [TNF]-alpha), and cytotoxic factor (granzyme B and perforin) mRNA expression. However, the drugs had different effect on Th2 type cytokines (IL-4 and IL-10). Whereas IL-4 expression was not affected by tacrolimus and was enhanced by CsA, IL-10 expression was more significantly suppressed by tacrolimus than CsA. Differences in the suppression of Th2 type cytokine gene expression indicate that the in vivo molecular networks by which CsA and tacrolimus exert their full immunosuppressive activity are not necessarily the same.
Collapse
Affiliation(s)
- H Jiang
- Fujisawa Research Institute of America, Evanston Northwestern Healthcare, Northwestern University/Evanston Research Park, IL 60201, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Krasinskas AM, Eiref SD, McLean AD, Kreisel D, Gelman AE, Popma SH, Moore JS, Rosengard BR. Replacement of graft-resident donor-type antigen presenting cells alters the tempo and pathogenesis of murine cardiac allograft rejection. Transplantation 2000; 70:514-21. [PMID: 10949196 DOI: 10.1097/00007890-200008150-00020] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Graft-resident antigen presenting cells (APCs) are potent stimulators of the alloresponse. To test whether replacement of graft-resident donor-type APCs with those of recipient-type alters allorecognition and the pathogenesis of both acute and chronic rejection, we created chimeric hearts for transplantation into naive recipients. METHODS To replace donor-type APCs with those of recipient-type, chimeric animals were created by bone marrow transplantation (BMT) in fully allogeneic mouse and rat strain combinations. The degree of APC replacement in chimeric organs was assessed phenotypically and functionally. Chimeric hearts were transplanted heterotopically into untreated recipients. RESULTS Flow cytometric and immunohistochemical analysis did not detect residual bone marrow recipient-type APCs in mouse BMT chimeras. Although semi-quantitative reverse transcription polymerase chain reaction detected 0.001-0.01% residual cells, APCs isolated from chimeric organs were functionally unable to stimulate donor-type cells. When transplanted into naive recipients, chimeric mouse hearts had significantly prolonged survival but were nevertheless rejected acutely. Similar results were obtained in the ACI --> LEW rat strain combination. However, in the PVG --> DA rat model, the majority of chimeric hearts survived >100 days and all long-surviving hearts developed cardiac allograft vasculopathy. CONCLUSIONS BMT leads to near complete replacement of organ-resident APCs. The virtual absence of donor-type APCs in chimeric hearts delays or prevents acute rejection in a strain-dependent manner. In contrast, this type of graft modification does not prevent cardiac allograft vasculopathy. This suggests that, although the CD4+ direct pathway may play a role in acute rejection, it is not essential for the development of chronic rejection in rodent cardiac allografts.
Collapse
Affiliation(s)
- A M Krasinskas
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Health System, Philadelphia 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Krieger NR, Fathman CG, Shaw MK, Ridgway WM. Identification and characterization of the antigen-specific subpopulation of alloreactive CD4+ T cells in vitro and in vivo. Transplantation 2000; 69:605-9. [PMID: 10708118 DOI: 10.1097/00007890-200002270-00023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We report the identification and characterization of the small subpopulation of alloantigen-specific T cells in vitro and in vivo. This subpopulation of T cells was distinguished by up-regulation of cell surface CD4 expression. These CD4high T cells were alloantigen specific in proliferation assays in vitro, and they expressed memory/activation markers, including CD44high and CD69high. Further studies demonstrated that these allospecific CD4high cells were also present (< or = 1% of CD4+ T cells) in vivo in BALB/c (H-2d) recipients of C57BL/6 (H-2b) skin allografts. CD4high T cells isolated from regional draining lymph nodes in these skin graft recipients reacted in a donor-specific fashion to C57BL/6 splenocyte stimulator cells in mixed lymphocyte culture. Adoptive transfer of CD4high, but not CD4normal T cells, just before skin engraftment in CD4 knockout mice, reconstituted rejection. The discovery that a small subpopulation of CD4high lymph node cells contained all of the alloantigen-specific T cells may allow study of tissue-specificity and subsequent alloantigen identification in transplantation.
Collapse
Affiliation(s)
- N R Krieger
- Department of Surgery, Stanford University, CA 94305-5111, USA
| | | | | | | |
Collapse
|
49
|
Haskova Z, Usiu N, Pepose JS, Ferguson TA, Stuart PM. CD4+ T cells are critical for corneal, but not skin, allograft rejection. Transplantation 2000; 69:483-7. [PMID: 10708099 DOI: 10.1097/00007890-200002270-00004] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The relative contribution of CD4+ or CD8+ T cells in allograft rejection remains to be fully characterized. Some reports indicate that there is an absolute requirement for CD4+ T cells in allogeneic rejection, whereas others report that CD4-depleted mice are capable of rejecting certain types of allografts. METHODS We compared the ability of CD4- knockout (KO), CD8- KO, and normal CD4+/CD8+ mice to reject allogeneic corneal or skin grafts. We also examined delayed-type hypersensitivity and CTL responses to donor alloantigens. RESULTS Engraftment of C57BL/6 corneas to C.B6-(n5-7) CD4-KO mice resulted in significantly higher rates of acceptance (>85%) than either C.B6-(n5-7) CD8- KO (30%) or normal BALB/c mice (40%). Likewise, mean survival times for B6 skin grafts placed on C.B6-(n5-7) CD4- KO mice (29.2 +/- 3.5 days) were significantly increased over those of normal BALB/c mice (13.2 +/- 1 days), although most CD4- KO mice (70%) eventually reject their grafts. C.B6-(n5-7) CD4- KO mice that reject allogeneic grafts fail to develop a delayed-type hypersensitivity response, but they did demonstrate significantly greater cytotoxic T lymphocyte precursor (CTLp) frequencies than did CD4- KO mice that accepted such grafts or that were not grafted. CONCLUSIONS This study indicates that mice lacking CD4+ T cells have a significantly impaired ability to reject corneal allografts, but are able, in most cases, to reject allogeneic skin grafts. Thus, in the absence of CD4+ T cells, the likely mechanism for rejection appears to involve the generation of CD8+ CTLs.
Collapse
Affiliation(s)
- Z Haskova
- Department of Ophthalmology and Visual Sciences, Washington University Medical School, St. Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
BACKGROUND Corneal transplantation is the oldest, most common, and arguably, the most successful form of tissue transplantation. In the United States alone, over 40,000 corneal transplantations are performed each year. Less than 10% of the uncomplicated, first-time corneal grafts will undergo immune rejection even though HLA matching is not routinely performed and the use of immunosuppressive drugs is limited to the topical application of corticosteroids. The success of corneal transplantations predates the use of corticosteroids and further emphasizes the remarkable privilege of corneal allografts. METHODS Several laboratories have used rat and mouse models of orthotopic corneal transplantation (keratoplasty) in an attempt to understand the basis for the immune privilege of corneal allografts. RESULTS The time-honored explanation for the immune privilege of corneal allografts was based on the conspicuous avascularity of the cornea, which was believed to sequester the graft from the immune apparatus. However, results from several laboratories indicate that at least three additional features of the corneal graft contribute to its immune privileged status: (a) absence of donor-derived, antigen-presenting passenger Langerhans cells in the corneal graft; (b) expression of Fas ligand on the epithelium and endothelium of the corneal allograft; and (c) capacity of the corneal allograft to induce immune deviation of the systemic immune response. CONCLUSIONS The immune privilege of corneal allografts is a product of at least three unique qualities of the corneal allograft that conspire to interfere with the induction and expression of allodestructive immune responses.
Collapse
Affiliation(s)
- J Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas 75235-9057, USA
| |
Collapse
|