151
|
Dufter C, Watzlik A, Christ C, Jung M, Wirzbach A, Opelz G, Terness P. Suppression of T-cell alloreactivity by gene-therapeutic modulation of human dendritic stimulator cells with TGF-beta adenoviral vectors. Transplant Proc 2001; 33:190-1. [PMID: 11266775 DOI: 10.1016/s0041-1345(00)01971-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- C Dufter
- Institute of Immunology, Department of Transplantation Immunology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
152
|
Mocchegiani E, Giacconi R, Muzzioli M, Cipriano C. Zinc, infections and immunosenescence. Mech Ageing Dev 2000; 121:21-35. [PMID: 11164457 PMCID: PMC7126297 DOI: 10.1016/s0047-6374(00)00194-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2000] [Revised: 07/25/2000] [Accepted: 08/12/2000] [Indexed: 10/25/2022]
Abstract
Infections may cause mortality in old age due to damaged immune responses. As zinc is required as a catalyst, structural (zinc fingers) and regulatory ion, it is involved in many biological functions, including immune responses. Low zinc ion bioavailability and impaired cell-mediated immunity are common in ageing and may be restored by physiological supplementation with zinc for 1-2 months, impacting upon morbidity and survival. This article reviews the role of zinc in immune efficacy during ageing, and also describes the main biochemical pathways involved in the role of zinc in resistance to infections in ageing in order to better understand the possible causes of immunosenescence.
Collapse
Affiliation(s)
- E Mocchegiani
- Immunology Centre (Section Nutrition, Immunity and Ageing) Research Department Nino Masera, Italian National Research Centres on Ageing (I.N.R.C.A.), Via Birarelli 8, 60121 Ancona, Italy.
| | | | | | | |
Collapse
|
153
|
Laub R, Dorsch M, Meyer D, Ermann J, Hedrich HJ, Emmrich F. A multiple transgenic mouse model with a partially humanized activation pathway for helper T cell responses. J Immunol Methods 2000; 246:37-50. [PMID: 11121545 DOI: 10.1016/s0022-1759(00)00288-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Mice expressing human CD4 and human MHC II molecules provide a valuable model both for the investigation of the immunopathogenetic role of human autoantigens and for the development of therapeutic strategies based on modulating helper T cell activation in vivo. Here we present a novel mouse model expressing HLA-DR17 (a split antigen of HLA-DR3) together with human CD4 in the absence of murine cd4 (CD4/DR3 mice). Human CD4 accurately replaces murine cd4 within T cells. In particular, the preservation of cd8(+) and CD4(+) T cell subsets distinguishes CD4/DR3 mice from other multiple transgenic models in which the alternative T cell subsets are fundamentally disturbed. Moreover, human CD4 is also faithfully expressed on antigen presenting cells such as dendritic cells and monocyte/macrophages, so that the overall transgenic CD4 expression pattern resembles very closely that of humans. HLA-DR3 expression in the thymus correlates very closely to that of mouse MHC II. In contrast, only 70% of mouse MHC II positive cells in spleen, lymph node, and peripheral blood coexpress HLA-DR3. No significant bias was found with regard to particular leucocytes in this respect. The stimulation of helper T cells clearly depends on the interaction between the human transgene products, since mAbs to HLA-DR and/or CD4 completely blocked in vitro recall responses to tetanus toxoid. CD4/DR3 mice represent a partially humanized animal model which will facilitate studies of DR3-associated autoimmune responses and the in vivo determination of the therapeutic potential of mAbs to human CD4.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- CD4 Antigens/biosynthesis
- CD4 Antigens/genetics
- CD4 Antigens/immunology
- CD8 Antigens/immunology
- Crosses, Genetic
- Down-Regulation
- Female
- Gene Expression
- HLA-DR3 Antigen/biosynthesis
- HLA-DR3 Antigen/genetics
- HLA-DR3 Antigen/immunology
- Humans
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lymphatic System/immunology
- Lymphatic System/metabolism
- Lymphocyte Activation/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Models, Animal
- Phenotype
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Tetanus Toxoid/immunology
- Transgenes
Collapse
Affiliation(s)
- R Laub
- Interdisciplinary Center of Clinical Research, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | |
Collapse
|
154
|
Jin Y, Fuller L, Wei Y, Blomberg BB, Miller J, Esquenazi V. Bone marrow cells promote TH2 polarization and inhibit virus-specific CTL generation. Hum Immunol 2000; 61:1233-41. [PMID: 11163078 DOI: 10.1016/s0198-8859(00)00206-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This laboratory recently reported that human bone marrow cells (BMC) inhibit the generation of virus-specific CTL in culture. The culture supernatants contained increased levels of prostaglandin E(2) (PGE(2)) (shown to favor TH2 cell development) and also inhibited EBV-CTL effector cell development. In this study, we obtained PBL from Epstein-Barr virus (EBV) IgG antibody positive kidney transplant recipients (R) and their living-related donors (LRD) one year after renal transplantation. EBV-specific CTL were then generated in vitro by stimulating PBL with autologous EBV-transformed B cells (EBV-B) in the presence or absence of autologous BMC. The addition of BMC to the EBV-CTL generation cultures increased the intracellular expression in CD3+ cells of IL-4,-5,-6,-10, and -13. These CD3+ cells also expressed increased levels of the TH2 associated receptor CCR3. Inhibition was even observed by preparing EBV-CTL generating cultures in trans-wells that separated the autologous BMC from the PBL + EBV-B. It was then observed that CD3+ cells obtained after 7 days of culture in the presence of autologous BMC could be used as inhibitors of EBV-CTL generation. Protein Kinase A (PKA), a cAMP kinase that is involved in the upregulation of TH2 cytokine activity, was increased in EBV-CTL cultures by the presence of BMC. Additionally, IL-4-mediated signal transduction and activation of transcription (STAT-6) phosphorylation was slightly increased. These results show that the BMC inhibition is mediated by soluble factors (cytokines) and that cell-cell contact in this autologous system is not required, so that BMC (at least partially, via cytokine production) promote TH2 polarization in culture. Moreover, TH2 cells induced by culturing with autologous BMC directly inhibit EBV-CTL generation, and TH2 associated PKA, CCR3, and STAT-6 phosphorylation are enhanced by BMC.
Collapse
Affiliation(s)
- Y Jin
- Division of Transplantation, Department of Surgery, University of Miami School of Medicine and the Miami Veterans Affairs Center, Miami, FL 33101, USA
| | | | | | | | | | | |
Collapse
|
155
|
Graca L, Honey K, Adams E, Cobbold SP, Waldmann H. Cutting edge: anti-CD154 therapeutic antibodies induce infectious transplantation tolerance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4783-6. [PMID: 11045999 DOI: 10.4049/jimmunol.165.9.4783] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Nondepleting anti-CD154 (CD40 ligand) mAbs have proven effective in inducing transplantation tolerance in rodents and primates. In the induction phase, anti-CD154 Ab therapy is known to enhance apoptosis of Ag reactive T cells. However, this may not be the sole explanation for tolerance, as we show in this study that tolerance is maintained through a dominant regulatory mechanism which, like tolerance induced with CD4 Abs, manifests as infectious tolerance. Therefore, tolerance induced with anti-CD154 Abs involves not only the deletion of potentially aggressive T cells, but also a contagious spread of tolerance to new cohorts of graft-reactive T cells as they arise.
Collapse
MESH Headings
- Adoptive Transfer
- Alemtuzumab
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/administration & dosage
- CD40 Ligand/immunology
- CD8-Positive T-Lymphocytes/immunology
- Injections, Intraperitoneal
- Injections, Intravenous
- Lymphocyte Depletion/methods
- Lymphocyte Transfusion
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Transgenic
- Skin Transplantation/immunology
- T-Lymphocytes/transplantation
- Thymectomy
- Transplantation Tolerance/genetics
- Transplantation Tolerance/immunology
Collapse
Affiliation(s)
- L Graca
- Sir William Dunn School of Pathology, Oxford, United Kingdom.
| | | | | | | | | |
Collapse
|
156
|
Foucras G, Coudert JD, Coureau C, Guéry JC. Dendritic cells prime in vivo alloreactive CD4 T lymphocytes toward type 2 cytokine- and TGF-beta-producing cells in the absence of CD8 T cell activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4994-5003. [PMID: 11046027 DOI: 10.4049/jimmunol.165.9.4994] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The mechanisms that influence the polarization of CD4 T cells specific for allogeneic MHC class II molecules in vivo are still poorly understood. We have examined the pathway of alloreactive CD4 T cell differentiation in a situation in which only CD4 T cells could be activated in vivo. In this report we show that priming of adult mice with allogeneic APC, in the absence of MHC class I-T cell interactions, induces a strong expansion of type 2 cytokine-producing allohelper T cells. These alloantigen-specific CD4 T cells directly recognize native allogeneic MHC class II molecules on APC and secrete, in addition to the prototypic Th2 cytokines IL-4, IL-5, and IL-10, large amounts of TGF-beta. The default Th2-phenotype acquisition is not genetically controlled and occurred both in BALB/c and C57BL/6 mice. CD8 T cells are the principal cell type that controls CD4 T cell differentiation in vivo. Furthermore, we demonstrate that strong Th2 priming can be induced not only with allogeneic splenocytes but also with a low number of bone marrow-derived dendritic cells. Finally, using a passive transfer system, we provide direct evidence that CD8 T cell expansion in situ promotes alloreactive Th1 cell development principally by preventing their default development to the Th2 pathway in a mechanism that is largely IFN-gamma independent. Therefore, this work demonstrates that type 2 cytokine production represents a dominant pathway of alloreactive CD4 T cell differentiation in adult mice, a phenomenon that was initially thought to occur only during the neonatal period.
Collapse
Affiliation(s)
- G Foucras
- Institut National de la Santé et de la Recherche Médicale Unité 28, Institut Fédératif de Recherche 30, Hôpital Purpan, Toulouse, France
| | | | | | | |
Collapse
|
157
|
Kedl RM, Rees WA, Hildeman DA, Schaefer B, Mitchell T, Kappler J, Marrack P. T cells compete for access to antigen-bearing antigen-presenting cells. J Exp Med 2000; 192:1105-13. [PMID: 11034600 PMCID: PMC2195874 DOI: 10.1084/jem.192.8.1105] [Citation(s) in RCA: 348] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2000] [Accepted: 07/24/2000] [Indexed: 11/04/2022] Open
Abstract
These studies tested whether antigenic competition between T cells occurs. We generated CD8(+) T cell responses in H-2(b) mice against the dominant ovalbumin epitope SIINFEKL (ova8) and subdominant epitope KRVVFDKL, using either vaccinia virus expressing ovalbumin (VV-ova) or peptide-pulsed dendritic cells. CD8(+) T cell responses were visualized by major histocompatibility complex class I-peptide tetrameric molecules. Transfer of transgenic T cells with high affinity for ova8 (OT1 T cells) completely inhibited the response of host antigen-specific T cells to either antigen, demonstrating that T cells can directly compete with each other for response to antigen. OT1 cells also inhibited CD8(+) T cell responses to an unrelated peptide, SIYRYGGL, providing it was presented on the same dendritic cells as ova8. These inhibitions were not due to a more rapid clearance of virus or antigen-presenting cells (APCs) by the OT1 cells. Rather, the inhibition was caused by competition for antigen and antigen-bearing cells, since it could be overcome by the injection of large numbers of antigen-pulsed dendritic cells. These results imply that common properties of T cell responses, such as epitope dominance and secondary response affinity maturation, are the result of competitive interactions between antigen-bearing APC and T cell subsets.
Collapse
Affiliation(s)
- R M Kedl
- Cancer Research Institute, National Jewish Medical and Research Center Denver, Colorado 80206, USA.
| | | | | | | | | | | | | |
Collapse
|
158
|
Lombardi G, Dunne PJ, Scheel-Toellner D, Sanyal T, Pilling D, Taams LS, Life P, Lord JM, Salmon M, Akbar AN. Type 1 IFN maintains the survival of anergic CD4+ T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:3782-9. [PMID: 11034383 DOI: 10.4049/jimmunol.165.7.3782] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anergic T cells have immunoregulatory activity and can survive for extended periods in vivo. It is unclear how anergic T cells escape from deletion, because both anergy and apoptosis can occur after TCR ligation. Stimulation of human CD4+ T cell clones reactive to influenza hemagglutinin peptides can occur in the absence of APCs when MHC class II-expressing, activated T cells present peptide to each other. This T:T peptide presentation can induce CD95-mediated apoptosis, while the cells that do not die are anergic. We found that the death after peptide or anti-CD3 treatment of a panel of CD4+ T cell clones is blocked by IFN-beta secreted by fibroblasts and also by IFN-alpha. This increases cell recovery after stimulation, which is not due to T cell proliferation. This mechanism for apoptosis inhibition rapidly stops protein kinase C-delta translocation from the cytoplasm to the nucleus, which is an early event in the death process. A central observation was that CD4+ T cells that are rescued from apoptosis after T:T presentation of peptide by IFN-alphabeta remain profoundly anergic to rechallenge with Ag-pulsed APCs. However, anergized cells retain the ability to respond to IL-2, showing that they are nonresponsive but functional. The prevention of peptide-induced apoptosis in activated T cells by IFN-alphabeta is a novel mechanism that may enable the survival and maintenance of anergic T cell populations after TCR engagement. This has important implications for the persistence of anergic T cells with the potential for immunoregulatory function in vivo.
Collapse
Affiliation(s)
- G Lombardi
- Department of Clinical Immunology, Royal Free and University College Medical School, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Abstract
Rejection of fully MHC-mismatched allografts entails the direct recognition of donor MHC molecules (direct antigen presentation) and the activation of an unusually large mass of alloreactive T cells. There is compelling evidence that apoptotic cell death of activated T cells is a critical initial step in the induction of peripheral allograft tolerance with regimens that are not inherently lymphoablative and that therapies that block T cell activation and T cell apoptosis also block the acquisition of tolerance. Thus, T cell apoptosis may play an important role in reducing the size of cytopathic T cell clones and this process may also promote the development and expansion of immune regulatory cells that are essential in the maintenance of allograft tolerance.
Collapse
Affiliation(s)
- X C Li
- Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
160
|
Gorczynski RM, Chen Z, Kai Y, Lei J. Evidence for persistent expression of OX2 as a necessary component of prolonged renal allograft survival following portal vein immunization. Clin Immunol 2000; 97:69-78. [PMID: 10998319 DOI: 10.1006/clim.2000.4907] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Following portal vein (pv) pretransplant immunization of C3H mice, there is an early (within 2 days) increase in expression of the molecule OX2 seen on host dendritic cells (DC), along with increased survival of C57BL/6 renal allografts transplanted within 24 h of pv immunization. In addition, there is a marked polarization in cytokine production from lymphocytes harvested from the transplanted animals, with preferential production of IL-4, IL-10, and TGFbeta on donor-specific restimulation in vitro, and decreased production of IL-2, IFN-gamma, and TNFalpha compared with non-pv-immunized control transplanted mice. Both the increased renal allograft survival and the altered cytokine production are abolished by infusion of anti-mouse OX2 monoclonal antibody (3B6), even when antibody infusion is begun as late as 10 days following transplantation. Quantitative PCR analysis independently shows that OX2 expression is increased in the spleen and liver of transplanted mice as late as 21 days following pv immunization. In vitro studies with an OX2:Fc immunoadhesion had suggested that immunosuppression induced by this soluble form of the OX2 molecule was dependent primarily upon an early (OX2-dependent) signal. This discrepancy between in vivo and in vitro data possibly reflects a role for OX2 in the in vivo recruitment of other (immunregulatory) cells. Consistent with this hypothesis, regardless of the time (posttransplantation) of in vivo infusion of anti-OX2 antibody, within 2 days we observed a decline in the functional activity of a previously characterized immunoregulatory gammadeltaTCR(+) cell population, which can be monitored by its ability to regulate cytokine production in vitro.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Graft Survival/drug effects
- Graft Survival/immunology
- Immunization
- Kidney Transplantation/immunology
- Liver/chemistry
- Male
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Orexin Receptors
- Portal Vein/immunology
- RNA, Messenger/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/analysis
- Receptors, G-Protein-Coupled
- Receptors, Neuropeptide/genetics
- Receptors, Neuropeptide/immunology
- Spleen/chemistry
- T-Lymphocytes, Regulatory/chemistry
- T-Lymphocytes, Regulatory/physiology
Collapse
|
161
|
Phillips JM, Harach SZ, Parish NM, Fehervari Z, Haskins K, Cooke A. Nondepleting anti-CD4 has an immediate action on diabetogenic effector cells, halting their destruction of pancreatic beta cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:1949-55. [PMID: 10925277 DOI: 10.4049/jimmunol.165.4.1949] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The induction of tolerance in a primed immune system is a major aim for therapy in autoimmunity and transplant rejection. In this paper, we investigate the action of the nondepleting anti-CD4 Ab, YTS 177. Although this Ab is nondepleting, we have demonstrated a direct action in vivo on activated effector cells. We show that the Ab inhibits transfer of insulin-dependent diabetes mellitus by the CD4+ Th1 clone BDC2.5 to nonobese diabetic mice. Furthermore, we show that this Ab acts directly on diabetogenic effector cells because it prevented BDC2.5-induced insulin-dependent diabetes mellitus in nonobese diabetic-scid recipients in the absence of other T cells. The Ab halts the diabetic process even when it is administered after the BDC2.5 cells have infiltrated the pancreas and destruction of islets is already underway. This is accompanied by an immediate decrease in proinflammatory cytokine production with cessation of beta cell destruction and disappearance of infiltrating cells from the pancreas, leaving any remaining beta cells intact. These data suggest that Abs such as this may be effective not only because they induce regulatory T cells but also because they are able to directly prevent effector cell function.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Animals, Newborn/immunology
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/pharmacology
- CD4 Antigens/immunology
- Cell Movement/immunology
- Clone Cells/immunology
- Clone Cells/transplantation
- Cytotoxicity, Immunologic/immunology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Enzyme Inhibitors/administration & dosage
- Enzyme Inhibitors/pharmacology
- Epithelium/immunology
- Epithelium/metabolism
- Female
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/pharmacology
- Histocompatibility Antigens Class II/biosynthesis
- Immunosuppressive Agents/administration & dosage
- Immunosuppressive Agents/pharmacology
- Injections, Intraperitoneal
- Interferon-gamma/antagonists & inhibitors
- Interferon-gamma/biosynthesis
- Interferon-gamma/genetics
- Interleukin-2/antagonists & inhibitors
- Interleukin-2/biosynthesis
- Interleukin-2/genetics
- Islets of Langerhans/enzymology
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Lymphocyte Activation/immunology
- Lymphocyte Depletion
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Nitric Oxide Synthase/antagonists & inhibitors
- Nitric Oxide Synthase/biosynthesis
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase Type II
- Pancreatic Ducts/immunology
- Pancreatic Ducts/metabolism
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/biosynthesis
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- J M Phillips
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
| | | | | | | | | | | |
Collapse
|
162
|
|
163
|
Zhang ZX, Yang L, Young KJ, DuTemple B, Zhang L. Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression. Nat Med 2000; 6:782-9. [PMID: 10888927 DOI: 10.1038/77513] [Citation(s) in RCA: 350] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite increasing evidence for the existence of antigen-specific regulatory T cells, the mechanisms underlying suppression remain unclear. In this study we have identified and cloned a novel subset of antigen-specific regulatory T cells and demonstrated that these T cells possess a unique combination of cell surface markers and array of cytokines. The regulatory T cells are able to inhibit the function of T cells carrying the same T-cell receptor specificity and prevent skin allograft rejection in an antigen-specific, dose-dependent manner. The regulatory T cells are able to acquire alloantigen from antigen-presenting cells, present the alloantigen to activated syngeneic CD8+ T cells and then send death signals to CD8+ T cells. These findings provide a novel mechanism of regulatory T-cell-mediated, antigen-specific suppression.
Collapse
Affiliation(s)
- Z X Zhang
- Department of Laboratory of Medicine and Pathobiology, Multi Organ Transplantation Program, Toronto General Hospital Research Institute, UHN, University of Toronto, Toronto, M5G, 2C4, Canada.
| | | | | | | | | |
Collapse
|
164
|
Hodgetts SI, Beilharz MW, Scalzo AA, Grounds MD. Why do cultured transplanted myoblasts die in vivo? DNA quantification shows enhanced survival of donor male myoblasts in host mice depleted of CD4+ and CD8+ cells or Nk1.1+ cells. Cell Transplant 2000; 9:489-502. [PMID: 11038066 DOI: 10.1177/096368970000900406] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Overcoming the massive and rapid death of injected donor myoblasts is the primary hurdle for successful myoblast transfer therapy (MTT), designed as a treatment for the lethal childhood myopathy Duchenne muscular dystrophy. The injection of male myoblasts into female host mice and quantification of surviving male DNA using the Y-chromosome-specific (Y1) probe allows the speed and extent of death of donor myoblasts to be determined. Cultured normal C57BL/10Sn male donor myoblasts were injected into untreated normal C57BL/10Sn and dystrophic mdx female host mice and analyzed by slot blots using a 32P-labeled Y1 probe. The amount of male DNA from donor myoblasts showed a remarkable decrease within minutes and by 1 h represented only about 10-18% of the 2.5 x 10(5) cells originally injected (designated 100%). This declined further over 1 week to approximately 1-4%. The host environment (normal or dystrophic) as well as the extent of passaging in tissue culture (early "P3" or late "P15-20" passage) made no difference to this result. Modulation of the host response by CD4+/CD8+ -depleting antibodies administered prior to injection of the cultured myoblasts dramatically enhanced donor myoblast survival in dystrophic mdx hosts (15-fold relative to untreated hosts after 1 week). NK1.1 depletion also dramatically enhanced donor myoblast survival in dystrophic mdx hosts (21-fold after 1 week) compared to untreated hosts. These results provide a strategic approach to enhance donor myoblast survival in clinical trials of MTT.
Collapse
Affiliation(s)
- S I Hodgetts
- Department of Anatomy and Human Biology, The University of Western Australia, Nedlands, Perth.
| | | | | | | |
Collapse
|
165
|
Yoo-Ott KA, Schiller H, Fändrich F, Oswald H, Richter K, Xhu XF, Kampen WU, Krönke M, Zavazava N. Co-transplantation of donor-derived hepatocytes induces long-term tolerance to cardiac allografts in a rat model. Transplantation 2000; 69:2538-46. [PMID: 10910274 DOI: 10.1097/00007890-200006270-00012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Liver allografts transplanted between MHC-disparate mice, rats, and swine are spontaneously accepted in most strain combinations without requirement for immunosuppression. The underlying mechanism has, however, remained elusive. Here, we demonstrate that co-transplantation of donor-derived hepatocytes protect Lewis (RT1.A1) cardiac allografts from acute and chronic rejection in DA (RT1.Aa) recipients indefinitely. METHODS Livers of donor Lewis rats were harvested and the hepatocytes separated from hepatic leukocytes by collagenase digestion and gradient separation. DA recipient animals were transplanted Lewis cardiac allografts and simultaneously intraportally infused either Lewis-derived hepatocytes or hepatic leukocytes. Recipient animals were either not further treated or received a single dose of 15 mg/kg cyclosporine. RESULTS Donor hepatocytes alone significantly protected syngeneic cardiac allografts from rejection, whereas hepatic leukocytes failed to influence graft survival. In combination with cyclosporine, recipient cardiac allografts were indefinitely protected from rejection. Graft-infiltrating cells in tolerant animals presented as clusters of CD4+ T cells and stained mostly positive for interleukin-4, whereas graft-infiltrating cells in rejected allografts were predominantly positive for interferon-gamma. Adoptive transfer of splenocytes derived from tolerant animals protected Lewis cardiac allografts from rejection in DA recipients without immunosuppression. In contrast, hepatic leukocytes protected only 50% of the allografts from rejection. CONCLUSION We propose that donor hepatocytes induce permanent engraftment of syngeneic allografts by establishing a Th2 type alloresponse that is transferable to new graft recipients. The results of this study demonstrate that liver parenchymal cells significantly mediate spontaneously liver-induced tolerance.
Collapse
Affiliation(s)
- K A Yoo-Ott
- Institute of Immunology, Department of General and Thoracic Surgery, University of Kiel, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
166
|
|
167
|
Santin AD, Hermonat PL, Ravaggi A, Bellone S, Pecorelli S, Roman JJ, Parham GP, Cannon MJ. Interleukin-10 increases Th1 cytokine production and cytotoxic potential in human papillomavirus-specific CD8(+) cytotoxic T lymphocytes. J Virol 2000; 74:4729-37. [PMID: 10775611 PMCID: PMC111995 DOI: 10.1128/jvi.74.10.4729-4737.2000] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin-10 (IL-10) is widely known as an immunosuppressive cytokine by virtue of its ability to inhibit macrophage-dependent antigen presentation, T-cell proliferation, and Th1 cytokine secretion. However, several studies have challenged the perception of IL-10 solely as an immunosuppressive cytokine. As part of an investigation on potentiation of the cytotoxic activity of human papillomavirus E7-specific CD8(+) cytotoxic T lymphocytes (CTL) for adoptive transfusions to cervical cancer patients, we found that IL-10 in combination with IL-2, unlike several other combinations, including IL-2 with IL-12, gamma interferon (IFN-gamma), tumor necrosis factor alpha, and transforming growth factor beta, was able to consistently increase cytotoxicity. This augmentation in cytotoxic activity correlated with a significant increase in the cytoplasmic accumulation of perforin as detected by fluorescence-activated cell sorter. Surface expression of both the alpha and beta chains of the CD8 heterodimeric coreceptor and CD56 molecules was increased by exposure of CTL to IL-10. More importantly, we found that administration of IL-10 in combination with IL-2 after antigen stimulation consistently increased the intracellular expression of Th1 cytokines (i.e., IFN-gamma and IL-2) compared to results for control CD8(+) T cells cultured in IL-2 alone. In kinetic studies, proliferation, intracellular perforin levels, cytotoxic activity, and IFN-gamma expression were consistently elevated in CTL cultures containing IL-10 compared to control cultures, both at early and late time points following stimulation. In contrast, intracellular IL-2 expression was consistently increased only at early time points following stimulation with autologous tumor cells or solid-phase anti-CD3 antibody. Taken together, these data support the use of IL-10 in combination with IL-2 for the in vitro expansion and potentiation of tumor-specific CTL for clinical use in the therapy of cancer.
Collapse
Affiliation(s)
- A D Santin
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205-7199, USA
| | | | | | | | | | | | | | | |
Collapse
|
168
|
Doetze A, Satoguina J, Burchard G, Rau T, Löliger C, Fleischer B, Hoerauf A. Antigen-specific cellular hyporesponsiveness in a chronic human helminth infection is mediated by T(h)3/T(r)1-type cytokines IL-10 and transforming growth factor-beta but not by a T(h)1 to T(h)2 shift. Int Immunol 2000; 12:623-30. [PMID: 10784608 DOI: 10.1093/intimm/12.5.623] [Citation(s) in RCA: 260] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Exposure to infective larvae of the filarial nematode Onchocerca volvulus (Ov) either results in patent infection (microfilaridermia) or it leads to a status called putative immunity, characterized by resistance to infection. Similar to other chronic helminth infections, there is a T cell proliferative hyporesponsiveness to Ov antigen (OvAg) by peripheral blood mononuclear cells (PBMC) from individuals with patent infection, i.e. generalized onchocerciasis (GEO), compared to PBMC from putatively immune (PI) individuals. In this study, mechanisms mediating this cellular hyporesponsiveness in GEO were investigated: the low proliferative response in PBMC from GEO individuals was associated with a lack of IL-4 production and significantly lower production of IL-5 compared to those from PI individuals, arguing against a general shift towards a T(h)2 response being the cause of hyporesponsiveness. In contrast, IL-10 and transforming growth factor (TGF)-beta, two cytokines associated with a T(h)3 response, seemed to mediate hyporesponsiveness: PBMC from individuals with GEO produced significantly more IL-10, and T cell proliferative hyporesponsiveness in this group could be reversed by the addition of anti-IL-10 and anti-TGF-beta antibodies. Hyporesponsiveness was specific for OvAg and not observed upon stimulation with related nematode antigens, arguing for a T cell-mediated, Ov-specific down-regulation. Ov-specific T cells could be cloned from GEO PBMC which have a unique cytokine profile (no IL-2 but high IL-10 and/or TGF-beta production), similar to the T cell subsets known to suppress ongoing inflammation (T(h)3 and T(r)1), indicating that this cell type which has not been found so far in infectious diseases may be involved in maintaining Ov-specific hyporesponsiveness.
Collapse
Affiliation(s)
- A Doetze
- Bernhard Nocht Institute of Tropical Medicine, 20359 Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
169
|
Hsu LJ, Jan MS, Lin YS. In vivo staphylococcal enterotoxin B (SEB)-primed murine splenocytes secrete mediators which suppress CD25(hi) expression and cell cycle progression of naive splenocytes in response to SEB in vitro. Cell Immunol 2000; 201:50-7. [PMID: 10805973 DOI: 10.1006/cimm.2000.1628] [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: 11/22/2022]
Abstract
Administration of bacterial superantigen results in clonal activation of T cells followed by a state of hyporesponsiveness to subsequent antigen stimulation. Using a coculture system, we showed that the splenocytes from staphylococcal enterotoxin B (SEB)-injected BALB/c mice suppressed the proliferative response of naive splenocytes to SEB stimulation. The suppressive effect also occurred in Fas-deficient MRL-lpr/lpr mice. When naive responder cells were separated by a semipermeable membrane from SEB-primed effector cells, the suppressive effect remained apparent. The hyporesponsiveness of responder cells did not result from excessive induction of apoptosis, but rather from prevention of entering the S and G2/M phases of the cell cycle. The IL-2 levels in culture supernatants were low with the presence of SEB-primed effector cells. However, addition of IL-2 to the cocultures only partially reversed the inhibitory effect. Further studies revealed a reduced level of the CD25(hi) subpopulation in responder cells when cultured in the transwell with the presence of SEB-primed effector cells compared to that with saline-primed controls. This inhibitory effect was not observed for SEB-induced activation of CD25(int) and CD69 expression. Taken together, using a transwell culture system, we show in this study an inhibition of CD25(hi) expression and cell cycle arrest in target cells, which may serve at least in part the mechanisms of SEB-induced hyporesponsiveness.
Collapse
Affiliation(s)
- L J Hsu
- Department of Microbiology and Immunology, National Cheng Kung University Medical College, Tainan, Taiwan, Republic of China
| | | | | |
Collapse
|
170
|
Dean GS, Tyrrell-Price J, Crawley E, Isenberg DA. Cytokines and systemic lupus erythematosus. Ann Rheum Dis 2000; 59:243-51. [PMID: 10733469 PMCID: PMC1753117 DOI: 10.1136/ard.59.4.243] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- G S Dean
- Centre for Rheumatology/ Bloomsbury Rheumatology Unit, Department of Medicine, University College, London, UK
| | | | | | | |
Collapse
|
171
|
|
172
|
Nashold FE, Miller DJ, Hayes CE. 1,25-dihydroxyvitamin D3 treatment decreases macrophage accumulation in the CNS of mice with experimental autoimmune encephalomyelitis. J Neuroimmunol 2000; 103:171-9. [PMID: 10696912 DOI: 10.1016/s0165-5728(99)00247-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sunlight, which is required for vitamin D biosynthesis, may be protective in multiple sclerosis (MS), due to the immunoregulatory functions of 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3), the hormonally active vitamin D metabolite. This hypothesis provided the impetus for the experiments reported here investigating mechanisms whereby 1,25-(OH)2D3 may inhibit murine experimental autoimmune encephalomyelitis (EAE). Severe EAE was induced, 1,25-(OH)2D3 or mock treatment was administered, and clinical disease, histopathological disease, and encephalitogenic cells in the central nervous system (CNS) were analyzed within 24-72 h of the treatment. The mock-treated mice remained paralyzed (stage 3 EAE) while most hormone-treated animals regained the partial use of both hind limbs (stage 2 EAE) within 72 h of treatment. A histopathological examination showed the hormone-treated mice had a 50% decrease in white matter and meningeal inflammation at 72 h post treatment. A flow cytometric analysis of cell surface markers on spinal cord cells recovered 24 h post treatment showed the mock-treated mice with EAE had about 7.0 +/- 2.3 million Mac-1+ cells/cord, whereas the hormone-treated mice had about 2.1 +/- 2.6 million Mac-1+ cells/cord, which was not significantly different from the unmanipulated control mice. Otherwise, the flow cytometric analysis detected no significant differences between the groups with respect to CD4+ or CD8+ T cells or B cells or macrophages in draining lymph nodes or spinal cords. These results are discussed with regard to possible fates for the 5 million Mac-1+ cells that were rapidly lost from the inflamed CNS in the 1,25-(OH)2D3-treated mice, and the possible beneficial effect of hormone treatment in resolving acute MS.
Collapse
Affiliation(s)
- F E Nashold
- Department of Biochemistry, University of Wisconsin-Madison, 53706, USA
| | | | | |
Collapse
|
173
|
Kurlberg G, Haglind E, Schön K, Törnqvist H, Lycke N. Blockade of the B7-CD28 pathway by CTLA4-Ig counteracts rejection and prolongs survival in small bowel transplantation. Scand J Immunol 2000; 51:224-30. [PMID: 10736090 DOI: 10.1046/j.1365-3083.2000.00725.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Allograft rejection involves T-cell activation, requiring T-cell receptor interactions with major histocompatibility complex (MHC) molecules and costimulatory signals delivered through the B7-CD28 pathway. We evaluated the effect of blocking this pathway on graft rejection and survival, in a rat experimental model of small bowel transplantation. Heterotopic small bowel transplantation was performed between PVG donor rats and DA recipient rats. The recipient animals were treated with CTLA4-Ig or irrelevant immunoglobulin (Ig)G as control and followed for 18, 30 or 90 days. The survival rate and degree of inflammation and accumulation of CD4+ T cells and macrophages were determined in the transplanted bowels. We found that administration of CTLA4-Ig significantly improved the survival rate compared to control rats: after 30 days 73% of the treated rats had survived and at 90 days 5/8 rats were still living, whereas in the control group only 2/8 rats had survived. The grafts showed preserved mucosal structure with only a mild degree of subacute inflammation and the accumulation of CD4+ T cells and macrophages was noticeably reduced in treated animals as compared to control rats. Necrosis was extensive in control rats, whereas CTLA4-Ig treated animals had grafts with at least some preserved villus morphology and no necrotic tissue. Although small bowel transplantation has proven exceptionally difficult, in this study we have shown that CTLA4-Ig treatment may provide a promising strategy to prevent rejection and induce long term tolerance and graft survival.
Collapse
Affiliation(s)
- G Kurlberg
- Departments of Surgery and Medical Microbiology and Immunology, University of Göteborg, S-413 45 Göteborg, Sweden
| | | | | | | | | |
Collapse
|
174
|
|
175
|
Choudhury A, Mukherjee P, Basu SK, George A, Rath S, Bal V. Disruption of T cell tolerance to self-immunoglobulin causes polyclonal B cell stimulation followed by inactivation of responding autoreactive T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:1713-21. [PMID: 10657615 DOI: 10.4049/jimmunol.164.4.1713] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Scavenger receptor (SR)-specific delivery by maleylation of a ubiquitous self-protein, Ig, to SR-bearing APCs results in self-limiting induction of autoimmune effects in vivo. Immunization with maleyl-Ig breaks T cell tolerance to self-Ig and causes hypergammaglobulinemia, with increases in spleen weight and cellularity. The majority of splenic B cells show an activated phenotype upon maleyl-Ig immunization, leading to large-scale conversion to a CD138+ phenotype and to significant increases in CD138-expressing splenic plasma cells. The polyclonal B cell activation, hypergammaglobulinemia, and autoreactive Ig-specific T cell responses decline over a 2-mo period postimmunization. Following adoptive transfer, T cells from maleyl-Ig-immune mice taken at 2 wk postimmunization can induce hypergammaglobulinemia in the recipients, but those taken at 10 wk postimmunization cannot. Hypergammaglobulinemia in the adoptive transfer recipients is also transient and is followed by an inability to respond to fresh maleyl-Ig immunization, suggesting that the autoreactive Ig-specific T cells are inactivated peripherally following disruption of tolerance. Thus, although autoreactive T cell responses to a ubiquitous self-Ag, Ig, are induced by SR-mediated delivery to professional APCs in vivo resulting in autoimmune pathophysiological effects, they are effectively and rapidly turned off by inactivation of these activated Ig-specific T cells in vivo.
Collapse
Affiliation(s)
- A Choudhury
- National Institute of Immunology, New Delhi, India
| | | | | | | | | | | |
Collapse
|
176
|
Abstract
Nonresponsiveness of mammalian T cells to self-antigens is not totally accounted for in terms of clonal deletion, T-cell anergy and T-cell ignorance: studies have shown that the T-cell repertoire of healthy individuals contains cells with the potential to cause autoimmune disease. This article describes a T-cell-mediated mechanism that prevents the realization of this potential and indicates how its failure can lead to the development of autoimmunity.
Collapse
Affiliation(s)
- B Seddon
- Division of Molecular Immunology, National Institute for Medical Research, The Ridgeway, Mill Hill, London, UK NW7 1AA
| | | |
Collapse
|
177
|
|
178
|
Segerer S, Nelson PJ, Schlöndorff D. Chemokines, chemokine receptors, and renal disease: from basic science to pathophysiologic and therapeutic studies. J Am Soc Nephrol 2000; 11:152-176. [PMID: 10616852 DOI: 10.1681/asn.v111152] [Citation(s) in RCA: 345] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Leukocyte trafficking from peripheral blood into affected tissues is an essential component of the inflammatory reaction to virtually all forms of injury and is an important factor in the development of many kidney diseases. Advances in the past few years have highlighted the central role of a family of chemotactic cytokines called chemokines in this process. Chemokines help to control the selective migration and activation of inflammatory cells into injured renal tissue. Chemokines and their receptors are expressed by intrinsic renal cells as well as by infiltrating cells during renal inflammation. This study summarizes the in vitro and in vivo data on chemokines and chemokine receptors in renal diseases with a special focus on potential therapeutic effects on inflammatory processes.
Collapse
Affiliation(s)
- Stephan Segerer
- Medizinische Poliklinik, Klinikum Innenstadt der Ludwig-Maximilians-Universität, Munich, Germany
| | - Peter J Nelson
- Medizinische Poliklinik, Klinikum Innenstadt der Ludwig-Maximilians-Universität, Munich, Germany
| | - Detlef Schlöndorff
- Medizinische Poliklinik, Klinikum Innenstadt der Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
179
|
Gao Q, Rouse TM, Kazmerzak K, Field EH. CD4+CD25+ cells regulate CD8 cell anergy in neonatal tolerant mice. Transplantation 1999; 68:1891-7. [PMID: 10628770 DOI: 10.1097/00007890-199912270-00013] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Injection of neonatal BALB/c mice with semi-allogeneic splenocytes leads to antigen-specific tolerance lasting into adulthood. Tolerant mice accept A/J skin grafts and fail to generate CD8 cytotoxic T lymphocyte (CTL) activity against A/J targets. Anergic CD8 T cells are present in tolerant mice, and CD4 regulatory cells function to maintain CD8 cell anergy. METHODS Neonatal BALB/c mice were injected with 108 live CAF, splenocytes, and mice were deemed tolerant by accepting A/J grafts over 40 days. CD8 cell proliferation was measured by in vitro incorporation of bromodeoxyuridine coupled with fluorescence-activated cell sorter analysis. Alloantigen-specific cytotoxicity was tested using 51Cr release assays of A/J or third-party targets. RESULTS We demonstrate that A/J-specific anergic CD8 cells are present in neonatal primed mice that develop tolerance but not in neonatal primed mice that reject A/J skin grafts. Anergic CD8 cells show decreased proliferation and no CTL activity against A/J targets. Addition of interleukin-2 (IL-2) to unfractionated cultures fails to restore CTL activity against A/J targets. However, addition of IL-2 to CD4-depleted cultures restores A/J-specific CD8 CTL activity. Removal of CD4+/CD25+ cells, but not CD4+/CD25- cells, also restores CD8 CTL activity against A/J in the presence, but not the absence, of IL-2. Moreover, when added back into cultures, purified CD4+/CD25+ cells from tolerant mice inhibit the generation of CD8 CTL against A/J targets. CONCLUSION These data indicate that CD8 anergy is associated with the state of tolerance, and that CD4+CD25+ cells from tolerant mice function to maintain A/J-specific CD8 cell anergy in vitro.
Collapse
Affiliation(s)
- Q Gao
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City 52242, USA
| | | | | | | |
Collapse
|
180
|
Abstract
Invertebrates use a nonadaptive, innate immunity, the expression of germline encoded receptors, to identify the allogeneic and xenogeneic attributes. Vertebrates also have the capacity to express ontogenically related adaptive immunity which is a somatically selected gene rearrangement process. Several commonly accepted generalizations are utilized to explain the enigmatic lack of the adaptive immunity in invertebrates. All point to the primitive nature of the innate immunity and the primitive organization of the body plan and the life history patterns of invertebrates. Seven of the most common generalizations are reviewed and confuted by virtue of a biased literature presentation. Subsequently, three evolutionary puzzles are raised and the accepted paradigm that the vertebrate immunity is pathogenically directed is further challenged. This leads to an alternative idea suggesting that preserving the individuality against the threat of invading conspecific cells might have been the original function of the immune system. This ancient system has been co-opted later on to serve as a defence mechanism against pathogens. The secondary role arose in the form of a multiplicity of newly developed phenomena, one of them being the vertebrate adaptive immunity. This proposal is supported by the fact that vertebrates still exhibit two distinct but common types of naturally occurring transplantation events (natural chimerism) and by a variety of recent studies, providing evidence for the crucial role of the vertebrate's innate immunity in signalling and triggering the acquired effector mechanisms.
Collapse
Affiliation(s)
- B Rinkevich
- National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, Haifa, Israel
| |
Collapse
|
181
|
Li Y, Li XC, Zheng XX, Wells AD, Turka LA, Strom TB. Blocking both signal 1 and signal 2 of T-cell activation prevents apoptosis of alloreactive T cells and induction of peripheral allograft tolerance. Nat Med 1999; 5:1298-302. [PMID: 10545997 DOI: 10.1038/15256] [Citation(s) in RCA: 597] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The alloimmune response against fully MHC-mismatched allografts, compared with immune responses to nominal antigens, entails an unusually large clonal size of alloreactive T cells. Thus, induction of peripheral allograft tolerance established in the absence of immune system ablation and reconstitution is a challenging task in transplantation. Here, we determined whether a reduction in the mass of alloreactive T cells due to apoptosis is an essential initial step for induction of stable allograft tolerance with non-lymphoablative therapy. Blocking both CD28-B7 and CD40-CD40 ligand interactions (co-stimulation blockade) inhibited proliferation of alloreactive T cells in vivo while allowing cell cycle-dependent T-cell apoptosis of proliferating T cells, with permanent engraftment of cardiac allografts but not skin allografts. Treatment with rapamycin plus co-stimulation blockade resulted in massive apoptosis of alloreactive T cells and produced stable skin allograft tolerance, a very stringent test of allograft tolerance. In contrast, treatment with cyclosporine A and co-stimulation blockade abolished T-cell proliferation and apoptosis, as well as the induction of stable allograft tolerance. Our data indicate that induction of T-cell apoptosis and peripheral allograft tolerance is prevented by blocking both signal 1 and signal 2 of T-cell activation.
Collapse
Affiliation(s)
- Y Li
- Department of Medicine, Harvard Medical School, Division of Immunology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | | | | | | | |
Collapse
|
182
|
Abstract
There has been considerable recent progress in the characterization of the regulatory T cells that mediate tolerance in a number of transplantation models. The conditions that facilitate the generation of regulatory T cells point to the thymus, the nature of immune suppression and the dose of immunosuppressive agent(s) being important. Putative mechanisms of immune regulation by regulatory T cells, particularly in the 'infectious' tolerance pathway, include Th2-type cytokines (IL-4, IL-10 and transforming growth factor beta) that may play a direct role as an indispensable requirement or may contribute indirectly as a favorable milieu for acquisition of tolerance. Anergic T cells may suppress immune responses via either cytokine competition or antigen-presenting cells. Models of autoimmune disease, in which regulatory T cells were shown to represent a distinct thymus-derived T cell subset, also suggest the role of antigen-presenting cells in mediating immune suppression. Progress has also been made in generating and characterizing regulatory T cells in vitro.
Collapse
Affiliation(s)
- Y Zhai
- The Dumont-UCLA Transplant Center, University of California (Los Angeles) School of Medicine, Department of Surgery, Room 77-120, Center for Health Science, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | | |
Collapse
|
183
|
Plebanski M, Flanagan KL, Lee EA, Reece WH, Hart K, Gelder C, Gillespie G, Pinder M, Hill AV. Interleukin 10-mediated immunosuppression by a variant CD4 T cell epitope of Plasmodium falciparum. Immunity 1999; 10:651-60. [PMID: 10403640 DOI: 10.1016/s1074-7613(00)80064-3] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The immunodominant CD4 T cell epitope region, Th2R, of the circumsporozoite protein of Plasmodium falciparum is highly polymorphic. Such variation might be utilized by the parasite to escape from or interfere with CD4 T cell effector functions. Here, we show that costimulation with naturally occurring altered peptide ligands (APL) can induce a rapid change from IFNgamma production to the immunosuppressive mediator interleukin 10 (IL-10). This mechanism may contribute to the low levels of T cell responses observed to this pathogen in malaria-endemic areas.
Collapse
Affiliation(s)
- M Plebanski
- Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Honey K, Cobbold SP, Waldmann H. Dominant regulation: a common mechanism of monoclonal antibody induced tolerance? Immunol Res 1999; 20:1-14. [PMID: 10467979 DOI: 10.1007/bf02786503] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Transplantation tolerance can be induced by a range of agents that block T cell/antigen-presenting cell (APC) interactions known to be important for initiation of the adaptive immune response. Tolerance so induced has been shown to have a regulatory phenotype dependent on CD4+ cells. This was first observed with nonlytic anti-CD4 antibodies, and was recently demonstrated following other therapeutic approaches. Dominant tolerance also plays a role in natural regulation of the immune response, functioning to prevent autoaggressive cells mediating self-destruction. The mechanism by which dominant tolerance is established and maintained remains unclear, and the reported characteristics of regulatory cells in different experimental models vary widely. Here we review the evidence for potential mechanisms involved and propose that there is a common pathway by which dominant tolerance is mediated.
Collapse
Affiliation(s)
- K Honey
- Sir William Dunn School of Pathology, Oxford, UK
| | | | | |
Collapse
|
185
|
Abstract
There is now compelling evidence that immune responses for both foreign and self antigens are downregulated by T cells that are specialised for this function; these are known as regulatory T (T reg) cells. This review describes progress in the characterisation of the T reg cells that mediate both mucosal tolerance and tolerance to self antigens. The recent work on the antigen specificity, generation and mode of action of T reg cells is also reviewed.
Collapse
Affiliation(s)
- D Mason
- MRC Cellular Immunology Unit Sir William Dunn School of Pathology University of Oxford Oxford OX1 3RE UK.
| | | |
Collapse
|