1
|
Rovito R, Claas FHJ, Haasnoot GW, Roelen DL, Kroes ACM, Eikmans M, Vossen ACTM. Congenital Cytomegalovirus Infection: Maternal-Child HLA-C, HLA-E, and HLA-G Affect Clinical Outcome. Front Immunol 2018; 8:1904. [PMID: 29354123 PMCID: PMC5760553 DOI: 10.3389/fimmu.2017.01904] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/13/2017] [Indexed: 01/02/2023] Open
Abstract
Congenital CMV infection (cCMV) is the most common congenital infection causing permanent long-term impairments (LTI). cCMV immunopathogenesis is largely unknown due to the complex interplay between viral, maternal, placental, and child factors. In this study, a large retrospective nationwide cohort of children with cCMV and their mothers was used. HLA-C, HLA-E, and HLA-G were assessed in 96 mother–child pairs in relation to symptoms at birth and LTI at 6 years of age. The mothers were additionally typed for killer cell immunoglobulin-like receptors. The maternal HLA-G 14 bp deletion/deletion polymorphism was associated with a worse outcome, as the immunomodulation effect of higher protein levels may induce less CMV control, with a direct impact on placenta and fetus. The absence of maternal HLA-C belonging to the C2 group was associated with symptoms at birth, as activating signals on decidual NK may override inhibitory signals, contributing to a placental pro-inflammatory environment. Here, the increased HLA-E*0101 and HLA-C mismatches, which were associated with symptoms at birth, may enhance maternal allo-reactivity to fetal Ags, and cause suboptimal viral clearance. Finally, HLA-C non-inherited maternal antigens (NIMAs) were associated with LTI. The tolerance induced in the fetus toward NIMAs may indirectly induce a suboptimal CMV antiviral response throughout childhood. In light of our findings, the potential role of maternal–child HLA in controlling CMV infection and cCMV-related disease, and the clinical value as predictor for long-term outcome certainly deserve further evaluation.
Collapse
Affiliation(s)
- Roberta Rovito
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Geert W Haasnoot
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Dave L Roelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Aloys C M Kroes
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - Michael Eikmans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Ann C T M Vossen
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
2
|
Tietze JK, Sckisel GD, Hsiao HH, Murphy WJ. Antigen-specific versus antigen-nonspecific immunotherapeutic approaches for human melanoma: the need for integration for optimal efficacy? Int Rev Immunol 2012; 30:238-93. [PMID: 22053969 DOI: 10.3109/08830185.2011.598977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Due to its immunogenecity and evidence of immune responses resulting in tumor regression, metastatic melanoma has been the target for numerous immunotherapeutic approaches. Unfortunately, based on the clinical outcomes, even the successful induction of tumor-specific responses does not correlate with efficacy. Immunotherapies can be divided into antigen-specific approaches, which seek to induce T cells specific to one or several known tumor associated antigens (TAA), or with antigen-nonspecific approaches, which generally activate T cells to become nonspecifically lytic effectors. Here the authors critically review the different immunotherapeutic approaches in melanoma.
Collapse
Affiliation(s)
- Julia K Tietze
- Departments of Dermatology and Internal Medicine, University of California-Davis, Sacramento, CA 95817, USA
| | | | | | | |
Collapse
|
3
|
Buschow SI, Nolte-'t Hoen ENM, van Niel G, Pols MS, ten Broeke T, Lauwen M, Ossendorp F, Melief CJM, Raposo G, Wubbolts R, Wauben MHM, Stoorvogel W. MHC II in dendritic cells is targeted to lysosomes or T cell-induced exosomes via distinct multivesicular body pathways. Traffic 2009; 10:1528-42. [PMID: 19682328 DOI: 10.1111/j.1600-0854.2009.00963.x] [Citation(s) in RCA: 322] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Dendritic cells (DCs) express major histocompatibility complex class II (MHC II) to present peptide antigens to T cells. In immature DCs, which bear low cell surface levels of MHC II, peptide-loaded MHC II is ubiquitinated. Ubiquitination drives the endocytosis and sorting of MHC II to the luminal vesicles of multivesicular bodies (MVBs) for lysosomal degradation. Ubiquitination of MHC II is abrogated in activated DCs, resulting in an increased cell surface expression. We here provide evidence for an alternative MVB sorting mechanism for MHC II in antigen-loaded DCs, which is triggered by cognately interacting antigen-specific CD4+ T cells. At these conditions, DCs generate MVBs with MHC II and CD9 carrying luminal vesicles that are secreted as exosomes and transferred to the interacting T cells. Sorting of MHC II into exosomes was, in contrast to lysosomal targeting, independent of MHC II ubiquitination but rather correlated with its incorporation into CD9 containing detergent-resistant membranes. Together, these data indicate two distinct MVB pathways: one for lysosomal targeting and the other for exosome secretion.
Collapse
Affiliation(s)
- Sonja I Buschow
- Faculty of Veterinary Medicine, Department of Biochemistry & Cell Biology, Utrecht University, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Kosec D, Vidić-Danković B, Isaković K, Leposavić G. ALTERATIONS IN THE THYMOPOIESIS IN EXPERIMENTAL AUTOIMMUNE MYASTHENIA GRAVIS. Int J Neurosci 2009; 115:461-77. [PMID: 15809214 DOI: 10.1080/00207450590522464] [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] [Indexed: 10/25/2022]
Abstract
Experimental autoimmune myasthenia gravis (EAGM) was induced in female AO rats, by a single immunization with Torpedo acetylcholine receptor (AChR). Animals injected with saline served as controls. The study showed substantial changes in EAMG rats in the thymopoiesis, causing an increase in the relative proportion of mature CD8+ and, particularly, CD4+ (possibly autoreactive) single positive (SP) cells expressing TCRalphabeta at high level (TCRalphabeta(high)), as well as in that of mature double negative (DN) TCRalphabeta(high) cells, which are believed to be the immunoregulatory cells that augment autoantibody (autoAb) production. These results indicate that an augmented production of autoreactive CD4+ cells, on one side, and an increase of the immunoregulatory T cells that augment autoAb production, on the other side (reflecting, most likely, an increased entry of activated autoreactive CD4+ T cells from the periphery into the thymus), may have a significant role in the sustention of immune response in EAMG, and may suggest a putative mechanism underlying the sustention of autoimmune response in acquired MG.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Female
- Flow Cytometry
- Lymphopoiesis/immunology
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Myasthenia Gravis, Autoimmune, Experimental/pathology
- Myasthenia Gravis, Autoimmune, Experimental/physiopathology
- Rats
- Rats, Inbred Strains
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Nicotinic/immunology
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/physiopathology
Collapse
Affiliation(s)
- Dusko Kosec
- Immunology Research Center Branislav Janković, Institute of Immunology and Virology Torlak, Belgrade, Serbia and Montenegro
| | | | | | | |
Collapse
|
5
|
Wang J, van Dongen H, Scherer HU, Huizinga TWJ, Toes REM. Suppressor activity among CD4+,CD25++ T cells is discriminated by membrane-bound tumor necrosis factor alpha. ACTA ACUST UNITED AC 2008; 58:1609-18. [PMID: 18512781 DOI: 10.1002/art.23460] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Previous studies have shown that the suppressive capacity of CD4+,CD25++ T cells is compromised in patients with rheumatoid arthritis (RA) and restored by anti-tumor necrosis factor alpha (anti-TNFalpha) therapy. Given the lack of specific cell surface markers for human Treg cells, this study aimed to define surface markers for identifying and enriching Treg cells with enhanced regulatory ability within the CD4+,CD25++ T cell compartment and to provide additional understanding of the effects of anti-TNFalpha antibodies in humans. METHODS The expression of membrane-bound TNFalpha in human peripheral blood CD4+ T cells was analyzed by flow cytometry in healthy individuals and RA patients before and after anti-TNFalpha treatment. Membrane-bound TNFalpha-positive and TNFalpha-negative CD4+,CD25++ T cells were purified by fluorescence-activated cell sorting, and their suppressive capacity was assessed in vitro by a standard suppression assay. RESULTS A substantial number of CD4+,CD25++ T cells expressed membrane-bound TNFalpha. Membrane-bound TNFalpha-positive CD4+,CD25++ T cells displayed reduced antiinflammatory cytokine production and less potent suppressor capacity, since 4 times more cells were required to achieve 50% inhibition compared with their membrane-bound TNFalpha-negative counterparts. Treatment of RA patients with TNFalpha-specific antibodies led to a reduction in the number of membrane-bound TNFalpha-positive CD4+,CD25++ T cells from peripheral blood. CONCLUSION Our data indicate that the absence of membrane-bound TNFalpha on CD4+,CD25++ T cells can be used to characterize and enrich for Treg cells with maximal suppressor potency. Enrichment of membrane-bound TNFalpha-negative CD4+,CD25+ cells in the CD4+,CD25++ T cell compartment may contribute to restoring the compromised suppressive ability of CD4+,CD25++ T cell populations in RA patients after anti-TNFalpha treatment.
Collapse
Affiliation(s)
- Jun Wang
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
6
|
Taams LS, Akbar AN. Peripheral generation and function of CD4+CD25+ regulatory T cells. Curr Top Microbiol Immunol 2005; 293:115-31. [PMID: 15981478 DOI: 10.1007/3-540-27702-1_6] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The balance between immunity and tolerance is important to maintain immune homeostasis. Several mechanisms are in place to ensure that the immune response is controlled, such as T cell anergy, apoptosis and immune ignorance. A fourth mechanism of peripheral tolerance is the active suppression by regulatory or suppressor T cells. The existence of suppressor T cells was first described in the early 1970s, but these cells became discredited in the 1980s. The work of Shimon Sakaguchi and others, however, has brought these cells back into the limelight and nowadays research into regulatory/suppressor T cells is a very active field of immunology. Different types of regulatory T cells have been described, including CD4+CD25+ T cells that constitutively express CTLA-4, GITR and Foxp3, TGF-beta producing Th3 cells, IL-10 producing Tr1 cells, and CD8+CD28- T cells. This review will focus on the generation and function of CD4+CD25+ regulatory T cells. CD4+CD25+ regulatory cells were originally described as thymus-derived anergic/suppressive T cells. Recent papers, however, indicate that these cells might also be generated in the periphery. CD4+CD25+ regulatory T cells can be activated by self-antigens and non-self-antigens, and once activated can suppress T cells in an antigen nonspecific manner. Interestingly, the suppressive effects of these cells are not restricted to the adaptive immune system (T and B cells) but can also affect the activation and function of innate immune cells (monocytes, macrophages, dendritic cells). These features make the CD4+CD25+ regulatory T cell subset an interesting target for immunotherapy of chronic inflammatory or autoimmune diseases.
Collapse
Affiliation(s)
- L S Taams
- Infection and Immunity Research Group, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NN, UK.
| | | |
Collapse
|
7
|
Waldmann H, Graca L, Adams E, Fairchild P, Cobbold S. Regulatory T cells in transplantation tolerance. Curr Top Microbiol Immunol 2005; 293:249-64. [PMID: 15981483 DOI: 10.1007/3-540-27702-1_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our ability to harness tolerance mechanisms will have a major impact in organ transplantation if it becomes possible to minimize drug maintenance, or even wean off immunosuppressive drugs. An improved understanding of the biology of regulatory T cells will make it possible to replace current induction regimens with those favouring the vaccination and selection of T cells that prevent graft rejection. Once tolerance is established, the continuous supply of graft antigens should sustain T cell mediated regulation as the dominant mechanism preventing graft rejection.
Collapse
Affiliation(s)
- H Waldmann
- Sir William Dunn School of Pathology, South Parks Road, Oxford OX1 3RE, UK.
| | | | | | | | | |
Collapse
|
8
|
Altin JG, van Broekhoven CL, Parish CR. Targeting dendritic cells with antigen-containing liposomes: antitumour immunity. Expert Opin Biol Ther 2005; 4:1735-47. [PMID: 15500402 DOI: 10.1517/14712598.4.11.1735] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Dendritic cells (DCs) are antigen-presenting cells that play an important role in the body's immune defence against cancer. Strategies using antigen-primed DCs as tumour vaccines show promise in patients, but the approach is cumbersome to use clinically. Soluble tumour antigens can be targeted to DCs in vivo, but this often induces antigenic tolerance rather than immunity. Liposomes are vesicular lipid structures with adjuvant-like properties. Importantly, liposomes can encapsulate antigen and immunomodulatory factors, thus serving as potent delivery vehicles. Different strategies are being explored to target liposomal antigens to DCs in vivo. One approach has employed single-chain antibody fragments to the DC surface molecules CD11c and DEC-205, attached to the vesicle surface by metal-chelating linkage, to target liposomal membranes containing antigen and either interferon-gamma or lipopolysaccharide to DCs. Such membranes induce dramatic antitumour responses and immunotherapeutic effects when used as a vaccine in the murine tumour model B16-OVA melanoma. Liposomal targeting of antigen and maturation signals directly to DCs in vivo, therefore, represents a much simpler strategy for cancer immunotherapy than antigen loading DCs ex vivo.
Collapse
Affiliation(s)
- Joseph G Altin
- The Australian National University, School of Biochemistry and Molecular Biology, Faculty of Science, Canberra, ACT 0200, Australia.
| | | | | |
Collapse
|
9
|
Abstract
Ever since the discovery of Medawar, over 50 years ago, that immunological tolerance was an acquired phenomenon that could be manipulated in neonatal mice, the ability to induce therapeutic tolerance against autoantigens, allergens and organ grafts has been a major driving force in immunology. Within the last 20 years we have found that a brief treatment with monoclonal antibodies that block certain functional molecules on the surface of the T cell is able to reprogramme the established immune repertoire of the adult mouse, allowing indefinite acceptance of allografts or effective curing of autoimmune diseases. We are only now just beginning to define many of the regulatory mechanisms that induce and maintain the tolerant state with the aim of being able to safely and reliably apply these technologies to human clinical situations.
Collapse
|
10
|
Kosec D, Lavrnić D, Apostolski S, Leposavić G. Changes in thymopoiesis in myasthenia gravis. Int J Neurosci 2005; 115:223-43. [PMID: 15764003 DOI: 10.1080/00207450590519472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This study was undertaken to investigate T-cell maturation in hyperplastic thymi of patients suffering from myasthenia gravis (MG). For this purpose, the expression of the major differentiational molecules (CD4, CD8, and CD3/TCRalphabeta) and that of the regulatory and activation molecules on thymocytes from MG patients and control subjects were estimated by flow cytometric analysis. In the MG patients the increase in relative proportion of immature (CD4-8- TCRalphabeta-) and the most mature (CD4+8- TCRalphabetahigh and CD4-8- TCRhigh encompassing immunoregulatory NKT) thymocytes followed by a decrease in that of CD4+8+CD3-/TCRalphabeta- cells was found. Furthermore, in these patients the relative proportion of CD4+HLA-DR+ and CD4+71+ cells was increased, whereas that of CD4+25+ cells was slightly, but significantly, decreased (reflecting, most likely, decreased contribution of T reg cells bearing this phenotype). Moreover, in MG thymi the percentage of CD45RA+ cells was reduced indicating changes in the selection processes. In keeping with this finding the reduced thymocyte apoptotic index and percentage of cells bearing apoptosing (CD4-8- TCRalphabetalow) phenotype were detected. In conclusion, the study demonstrates substantial changes in intrathymic differentiation of T cells in hyperplastic MG thymi and suggests alterations in selection events providing an increased escape of potentially autoreactive T-cell clones, on one side, and an altered maturation and/or selection of immunoregulatory cells (NKT and CD4+8-25+ T reg cells) keeping these cell clones under control, on the other side.
Collapse
MESH Headings
- Adult
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- CD4 Antigens/immunology
- CD4 Antigens/metabolism
- CD8 Antigens/immunology
- CD8 Antigens/metabolism
- Disease Progression
- Female
- Flow Cytometry
- Fluorescence
- HLA-DR Antigens/immunology
- HLA-DR Antigens/metabolism
- Humans
- Leukocyte Common Antigens/immunology
- Leukocyte Common Antigens/metabolism
- Myasthenia Gravis/immunology
- Myasthenia Gravis/metabolism
- Myasthenia Gravis/physiopathology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Thymopoietins/immunology
- Thymopoietins/metabolism
- Thymopoietins/physiology
Collapse
Affiliation(s)
- Dusko Kosec
- Immunology Research Center Branislav Jankovic, Institute of Immunology and Virology, TORLAK, Belgrade, Serbia and Montenegro
| | | | | | | |
Collapse
|
11
|
Nolte-'t Hoen ENM, Wagenaar-Hilbers JPA, Peters PJ, Gadella BM, van Eden W, Wauben MHM. Uptake of membrane molecules from T cells endows antigen-presenting cells with novel functional properties. Eur J Immunol 2004; 34:3115-25. [PMID: 15459903 DOI: 10.1002/eji.200324711] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although intercellular transfer of cell surface molecules has been observed between several cells of the immune system, the physiological relevance of this phenomenon remained obscure. Until now the transfer of molecules between antigen-presenting cells (APC) and T cells has been described as a unidirectional process from APC to T cells. However, here we show that T cells in turn donate molecules to APC, and that T cell-derived vesicles can mediate this transfer. The transferred proteins are incorporated into the APC as active molecules. Our data provide evidence that T cells use intercellular molecule transfer to mediate cell contact-dependent regulation of T cell responses via modulation of the APC.
Collapse
Affiliation(s)
- Esther N M Nolte-'t Hoen
- Department of Infectious Diseases and Immunology, Division of Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
12
|
Tang X, Yocum DE, Dejonghe D, Nordensson K, Lake DF, Richard J. Increased activation-induced cell death in peripheral lymphocytes of rheumatoid arthritis patients: the mechanism of action. Immunology 2004; 112:496-505. [PMID: 15196219 PMCID: PMC1782498 DOI: 10.1111/j.1365-2567.2004.01888.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Recently, we have described a soluble survival signal for activated lymphocytes from CD14(+) cells. As a result of the importance of T lymphocytes in the pathogenesis of rheumatoid arthritis (RA), we speculate a possible role for CD14(+) cells in supporting the outgrowth of autoreactive lymphocytes in RA. To address this issue further, supernatants from activated CD14(+) cells (CD14 cocktails) in both normal controls and RA patients were collected. The relative strength of the CD14 cocktails from normal controls and RA patients was compared. The data showed that depletion of CD14(+) cells resulted in a much higher increase of activation-induced cell death (AICD) and a decrease of lymphocyte proliferation in the peripheral blood mononuclear cells of RA patients compared to normal controls. Interestingly, CD14 cocktails from RA patients provide much stronger protection against AICD compared to those from normal controls. The observed soluble survival signal from CD14(+) cells is a general phenomenon because CD14 cocktails prevent both phytohaemagglutinin A-p- and anti-CD3-induced AICD. Furthermore, supernatants collected from human dendritic cell cultures also prevent activated lymphocytes from undergoing AICD. The data implicate an important role of the CD14(+) cell and its secreted form of survival signal in the pathogenesis of RA.
Collapse
Affiliation(s)
- Xiaolei Tang
- The Department of Microbiology and Immunology, the Arizona Arthritis Center, the University of Arizona, Tucson, AZ 85721, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Verhasselt V, Vosters O, Beuneu C, Nicaise C, Stordeur P, Goldman M. Induction of FOXP3-expressing regulatory CD4pos T cells by human mature autologous dendritic cells. Eur J Immunol 2004; 34:762-772. [PMID: 14991606 DOI: 10.1002/eji.200324552] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Current literature suggests that T cells recognizing antigen on mature dendritic cells (DC) differentiate into effector T cells whereas tolerance is induced when antigen is presented by immature DC. We investigated the consequences of the interactions between immature or lipopolysaccharide-matured DC and CD4(pos) T lymphocytes in absence of foreign antigen. While immature DC did not induce significant CD4(pos) T cell activation, we observed that a significant fraction of CD4(pos) T cells cultured with mature autologous DC displayed phenotypic features of activation and produced IL-2, IFN-gamma, IL-10 and TGF-beta. Furthermore, CD4(pos) T lymphocytes primed by mature, but not immature, autologous DC acquired regulatory properties. Indeed, when added to an allogeneic mixed leukocyte reaction, they suppressed the response of alloreactive T lymphocytes to the priming DC while responses to third-party stimulators were spared. The generation of CD4(pos) T cells with regulatory function by autologous stimulation did not require the presence of natural CD4(pos)CD25(pos) regulatory T cells. In addition, the acquisition of regulatory function by CD4(pos)CD25(neg) T cells stimulated by autologous mature DC was accompanied by the induction of FOXP3 expression. Our data suggest that during inflammatory conditions, presentation of self antigens by mature DC to autologous T lymphocytes could contribute to the generation of regulatory mechanisms.
Collapse
Affiliation(s)
- Valérie Verhasselt
- Laboratory of Experimental Immunology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- present address: INSERM-03 44, Université de Nice-Sophia Antipolis, IPMC, Valbonne, France
| | - Olivier Vosters
- Laboratory of Experimental Immunology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Claire Beuneu
- Laboratory of Experimental Immunology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Charles Nicaise
- Laboratory of Experimental Immunology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Patrick Stordeur
- Laboratory of Experimental Immunology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Michel Goldman
- Laboratory of Experimental Immunology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
14
|
Duthoit CT, Nguyen P, Geiger TL. Antigen Nonspecific Suppression of T Cell Responses by Activated Stimulation-Refractory CD4+ T Cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:2238-46. [PMID: 14764692 DOI: 10.4049/jimmunol.172.4.2238] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Several classes of anergic T cells are capable of suppressing naive T cell proliferation and thereby limiting immune responses. Activated T cells, although not anergic, are transiently refractory to restimulation with Ag. We examine in this study whether activated refractory murine T cells can also suppress naive T cell responses. We find that they can, and that they exhibit many of the suppressive properties of anergic T cells. The activated cells strongly diminish Ag-mediated T cell proliferation, an activity that correlates with their refractory period. Suppression is independent of APC numbers and requires cell contact or proximity. Naive T cells stimulated in the presence of activated refractory cells up-regulate CD25 and CD69, but fail to produce IL-2. The addition of IL-2 to culture medium, however, does not prevent the suppression, which is therefore not solely due to the absence of this growth factor. Persistence of the suppressor cells is also not essential. T cells stimulated in their presence and then isolated from them and cultured do not divide. The suppressive cells, however, do not confer a refractory or anergic state on the target T lymphocytes, which can fully respond to antigenic stimulation if removed from the suppressors. Our results therefore provide evidence that activated T cells act as transient suppressor cells, severely constraining bystander T cell stimulation and thereby restricting their response. These results have potentially broad implications for the development and regulation of immune responses.
Collapse
Affiliation(s)
- Christine T Duthoit
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | |
Collapse
|
15
|
Unger WWJ, Hauet-Broere F, Jansen W, van Berkel LA, Kraal G, Samsom JN. Early events in peripheral regulatory T cell induction via the nasal mucosa. THE JOURNAL OF IMMUNOLOGY 2004; 171:4592-603. [PMID: 14568933 DOI: 10.4049/jimmunol.171.9.4592] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Nasal application of soluble Ags leads to Ag-specific suppression of systemic immune responses. This tolerance can be transferred to naive mice by CD4(+) regulatory T cells (T(R) cells) from the spleen, but little is known about the induction of mucosal T(R) cells in vivo. To investigate the induction of T(R) cells in the nose-draining cervical lymph node (CLN), CD4(+) T cells from DO11.10 OVA TCR transgenic mice were transferred to BALB/c recipients. Within 48 h after nasal OVA application, CD4(+) DO11.10 T cells in CLN, but not in the peripheral lymph node, had divided. Similarly, nonmucosal (i.m.) OVA application also induced CD4(+) DO11.10 T cells to proliferate in the draining inguinal lymph node (ILN), yet more vigorously and with different kinetics than the CD4(+) DO11.10 T cells in CLN. Functional analysis revealed that only proliferating CD4(+) DO11.10 T cells from CLN, and not ILN, could transfer tolerance to naive recipients. CD4(+) DO11.10 T cells from CLN were phenotypically similar to CD4(+) DO11.10 T cells from ILN, however, in CLN a higher percentage of CD25(+) proliferating CD4(+) DO11.10 T cells were detected compared with ILN. CD25 is not a discriminative marker for mucosal T(R) cells because both CD25(+) and CD25(-) CD4(+) DO11.10 T cells from the CLN could suppress delayed type hypersensitivity responses in adoptive transfer. These findings demonstrate that although striking similarities exist between the differentiation of T(R) and effector T cells, this does not include their function. We are the first to demonstrate that functional T(R) cells, which reside within both CD25(+) and CD25(-) subsets, can be isolated from CLN as early as 3 days after nasal OVA application.
Collapse
MESH Headings
- ADP-ribosyl Cyclase/biosynthesis
- ADP-ribosyl Cyclase 1
- Administration, Intranasal
- Adoptive Transfer
- Animals
- Antigens, CD/biosynthesis
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CTLA-4 Antigen
- Cell Division/immunology
- Cell Separation
- Cytokines/metabolism
- Dose-Response Relationship, Drug
- Down-Regulation/immunology
- Female
- Hyaluronan Receptors/biosynthesis
- Immune Tolerance/immunology
- Immunophenotyping
- L-Selectin/biosynthesis
- Lectins, C-Type
- Leukocyte Common Antigens/biosynthesis
- Lipopolysaccharides/administration & dosage
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Membrane Glycoproteins
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Nasal Mucosa/cytology
- Nasal Mucosa/immunology
- Nasal Mucosa/metabolism
- Neck
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Receptors, Interleukin-2/biosynthesis
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- Time Factors
Collapse
Affiliation(s)
- Wendy W J Unger
- Department of Molecular Cell Biology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
16
|
Nakken B, Davis KE, Pan ZJ, Bachmann M, Farris AD. T-helper cell tolerance to ubiquitous nuclear antigens. Scand J Immunol 2003; 58:478-92. [PMID: 14629620 PMCID: PMC2579760 DOI: 10.1046/j.1365-3083.2003.01323.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Systemic autoimmune diseases are characterized by the development of antinuclear autoantibodies. In order to understand the immunologic events leading to the development of such antibodies, knowledge of mechanisms of immune tolerance to nuclear antigens is required. By utilizing adoptive T-cell transfer strategies with transgenic mouse models expressing nuclear neo-self antigens, T-cell tolerance to the lupus-related nuclear antigens human La and nRNP A has been demonstrated. These findings also indicate the existence in normal animals of autoreactive B cells continuously presenting nuclear antigen, suggesting that nuclear antigens are not sequestered from the immune system. Investigations of CD4+ T-cell tolerance to non-nuclear antigens have revealed a number of mechanisms that protect the host from autoreactivity, including autoreactive T-cell deletion, regulatory T-cell development and anergy induction. Recent studies using T-cell receptor and neo-self nuclear antigen transgenic mice are revealing the importance of such mechanisms in maintaining tolerance to nuclear antigens. Mechanisms of tolerogenic antigen presentation, identification of tolerogenic antigen source(s) and the pathways leading to loss of tolerance to nuclear antigens in systemic autoimmune disease states are currently being sought.
Collapse
Affiliation(s)
- B Nakken
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
17
|
Abstract
T cell anergy is a tolerance mechanism in which the lymphocyte is intrinsically functionally inactivated following an antigen encounter, but remains alive for an extended period of time in a hyporesponsive state. Models of T cell anergy affecting both CD4(+) and CD8(+) cells fall into two broad categories. One, clonal anergy, is principally a growth arrest state, whereas the other, adaptive tolerance or in vivo anergy, represents a more generalized inhibition of proliferation and effector functions. The former arises from incomplete T cell activation, is mostly observed in previously activated T cells, is maintained by a block in the Ras/MAP kinase pathway, can be reversed by IL-2 or anti-OX40 signaling, and usually does not result in the inhibition of effector functions. The latter is most often initiated in naïve T cells in vivo by stimulation in an environment deficient in costimulation or high in coinhibition. Adaptive tolerance can be induced in the thymus or in the periphery. The cells proliferate and differentiate to varying degrees and then downregulate both functions in the face of persistent antigen. The state involves an early block in tyrosine kinase activation, which predominantly inhibits calcium mobilization, and an independent mechanism that blocks signaling through the IL-2 receptor. Adaptive tolerance reverses in the absence of antigen. Aspects of both of the anergic states are found in regulatory T cells, possibly preventing them from dominating initial immune responses to foreign antigens and shutting down such responses prematurely.
Collapse
Affiliation(s)
- Ronald H Schwartz
- Laboratory of Cellular and Molecular Immunology, National Institutes of Health, Bethesda, Maryland 20892-0420, USA.
| |
Collapse
|
18
|
Bickerstaff A, Orosz C. Evidence for a limited contribution of immune regulation to cardiac allograft acceptance. Hum Immunol 2002; 63:935-47. [PMID: 12368046 DOI: 10.1016/s0198-8859(02)00447-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have used donor-reactive DTH responses to study the immune regulation that is displayed by C57BL/6 mice after they accept DBA/2 cardiac or renal allografts. This regulation is expressed by splenocytes from the allograft acceptors, and involves their alloantigen-induced production of the anti-inflammatory cytokines transforming growth factor beta (TGFbeta) and/or interleukin-10 (IL-10) at DTH challenge sites, and presumably within accepted allografts. In cardiac allograft acceptors the production of these cytokines depends on a population of CD25(+) splenocytes. During these studies, we have encountered several situations in which allograft acceptance does not correlate with DTH-detectable immune regulation: (1) splenocytes from cardiac or kidney allograft acceptors lose TGFbeta-mediated inhibition of donor-reactive DTH responses by 150 days post-transplant, although they retain ongoing allograft function; (2) cardiac allograft acceptors rapidly reject donor-matched skin allografts, retain good cardiac allograft function, but lose DTH-detectable immune regulation; (3) Balb/c mice accept C57BL/6 cardiac allografts when treated with anti-CD40L mAb (MR1), but fail to express DTH-detectable immune regulation; and (4) infusion of C57BL/6 mice with peritoneal exudate cells (PEC) that were educated ex vivo to DBA/2 alloantigens in the presence of IL-10 and TGFbeta, causes them to exhibit DTH-detectable immune regulation mediated by both TGFbeta and IL-10, but they fail to accept DBA/2 cardiac allografts. These observations suggest that the process of allograft acceptance, as it is studied in murine transplant models, is metastable, and does not necessarily reflect the achievement of allograft tolerance. Further, the development of allograft tolerance probably requires more than regulatory T cells, representing a coordinated evolution of multiple immune processes over a prolonged period of time.
Collapse
Affiliation(s)
- Alice Bickerstaff
- Department of Surgery, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | | |
Collapse
|
19
|
Papassavas AC, Stavropoulos-Giokas C, Boletis J, Ioannou S, Iniotaki-Theodoraki A, Kostakis A. Definition of permissible and immunogenic HLA antigens based on epitope analysis of the HLA specific antibodies produced in sensitized patients. EUROPEAN JOURNAL OF IMMUNOGENETICS : OFFICIAL JOURNAL OF THE BRITISH SOCIETY FOR HISTOCOMPATIBILITY AND IMMUNOGENETICS 2002; 29:401-7. [PMID: 12358849 DOI: 10.1046/j.1365-2370.2002.00341.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The goal of this study was to develop an accurate protocol whereby detection of acceptable HLA-A and -B mismatches is based on epitope analysis of HLA class I specific antibodies detected in the serum of highly sensitized patients awaiting a kidney retransplant. A total of 400 serum samples from 44 highly sensitized patients with panel reactive antibodies (PRA) of > or = 60% were collected during a 3-year follow-up period. All patients had been sensitized from a previous graft. In order to define the specificities of the HLA class I specific antibodies, two techniques were used in parallel: the antihuman globulin augmented complement-dependent cytotoxicity (CDC) technique and an enzyme-linked immunoabsorbent assay (ELISA) technique. Epitope identification was based on class I HLA antigen sequencing, where the unique epitope configuration on one HLA antigen represented the private epitope of the specific HLA antigen, and epitopes shared by more than one HLA antigen represented public determinants. The epitope prediction for the immunogenic HLA epitopes was based on an MHC database. For each highly sensitized patient, antibody specificities against actual and 'at risk' epitopes were defined. Following epitope analysis, all HLA antigens that did not express the actual and/or 'at risk' immunogenic epitopes were considered as acceptable mismatches of epitope analysis. The cytotoxicity of highly sensitized patients was determined using two different panels of selected, separated T lymphocytes. HLA class I specific IgG antibodies against 69 actual and 86 'at risk' epitopes were detected. In all patients, a large number of acceptable mismatches were defined. These included a large number of HLA antigens, corresponding to both HLA-A and -B loci. Our study introduces an accurate protocol for the detection of acceptable mismatches in highly sensitized patients. According to this protocol, the detailed description of immunogenic HLA specific epitope targets, against which HLA class I specific antibodies are directed, is a useful tool for the detection of acceptable mismatches in highly sensitized patients. This may lead to reduced production of HLA class I specific antibodies and, consequently, improved graft survival.
Collapse
Affiliation(s)
- A C Papassavas
- Department of Immunology and National Tissue Typing Center, General Hospital Georgios Gennimatas, Athens, Greece.
| | | | | | | | | | | |
Collapse
|
20
|
Zhong MC, Kerlero de Rosbo N, Ben-Nun A. Multiantigen/multiepitope-directed immune-specific suppression of "complex autoimmune encephalomyelitis" by a novel protein product of a synthetic gene. J Clin Invest 2002; 110:81-90. [PMID: 12093891 PMCID: PMC151033 DOI: 10.1172/jci15692] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Systemic administration of antigen/peptide for peripheral T cell tolerance has long been investigated as a potential approach to therapy of autoimmune diseases. The multiple antimyelin T cell reactivities likely to be associated with multiple sclerosis (MS) impose major difficulties in devising such an immune-specific therapeutic approach to the disease, because targeting T cells specific for a single autoantigen/epitope is unlikely to be sufficiently effective. Here, we present a pilot study on the possibility of concomitantly inhibiting multiple potentially pathogenic antimyelin T cell reactivities by tolerogenic administration of an artificial "multiantigen/multiepitope" protein. A synthetic gene was constructed to encode selected disease-relevant epitopes of myelin basic protein (MBP), proteolipid protein (PLP), and myelin oligodendrocyte glycoprotein (MOG). The protein product, hmTAP (synthetic human multitarget autoantigen protein), was adequately processed for antigenic presentation of the relevant integral epitopes, in vitro and in vivo. Systemic administration of hmTAP not only suppressed and treated experimental autoimmune encephalomyelitis (EAE) initiated by autoreactivity to a PLP epitope, but also abrogated complex EAE transferred by multispecific line T cells reactive against encephalitogenic epitopes of MBP, PLP, and MOG. These data indicate that multiantigen/multiepitope-directed therapy of complex autoimmune diseases is effective and can be mediated by the protein product of a specifically designed synthetic gene.
Collapse
Affiliation(s)
- Ming-Chao Zhong
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | |
Collapse
|
21
|
Burgess SC, Davison TF. Identification of the neoplastically transformed cells in Marek's disease herpesvirus-induced lymphomas: recognition by the monoclonal antibody AV37. J Virol 2002; 76:7276-92. [PMID: 12072527 PMCID: PMC136297 DOI: 10.1128/jvi.76.14.7276-7292.2002] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2002] [Accepted: 04/18/2002] [Indexed: 11/20/2022] Open
Abstract
Understanding the interactions between herpesviruses and their host cells and also the interactions between neoplastically transformed cells and the host immune system is fundamental to understanding the mechanisms of herpesvirus oncology. However, this has been difficult as no animal models of herpesvirus-induced oncogenesis in the natural host exist in which neoplastically transformed cells are also definitively identified and may be studied in vivo. Marek's disease (MD) herpesvirus (MDV) of poultry, although a recognized natural oncogenic virus causing T-cell lymphomas, is no exception. In this work, we identify for the first time the neoplastically transformed cells in MD as the CD4(+) major histocompatibility complex (MHC) class I(hi), MHC class II(hi), interleukin-2 receptor alpha-chain-positive, CD28(lo/-), phosphoprotein 38-negative (pp38(-)), glycoprotein B-negative (gB(-)), alphabeta T-cell-receptor-positive (TCR(+)) cells which uniquely overexpress a novel host-encoded extracellular antigen that is also expressed by MDV-transformed cell lines and recognized by the monoclonal antibody (MAb) AV37. Normal uninfected leukocytes and MD lymphoma cells were isolated directly ex vivo and examined by flow cytometry with MAb recognizing AV37, known leukocyte antigens, and MDV antigens pp38 and gB. CD28 mRNA was examined by PCR. Cell cycle distribution and in vitro survival were compared for each lymphoma cell population. We demonstrate for the first time that the antigen recognized by AV37 is expressed at very low levels by small minorities of uninfected leukocytes, whereas particular MD lymphoma cells uniquely express extremely high levels of the AV37 antigen; the AV37(hi) MD lymphoma cells fulfill the accepted criteria for neoplastic transformation in vivo (protection from cell death despite hyperproliferation, presence in all MD lymphomas, and not supportive of MDV production); the lymphoma environment is essential for AV37(+) MD lymphoma cell survival; pp38 is an antigen expressed during MDV-productive infection and is not expressed by neoplastically transformed cells in vivo; AV37(+) MD lymphoma cells have the putative immune evasion mechanism of CD28 down-regulation; AV37(hi) peripheral blood leukocytes appear early after MDV infection in both MD-resistant and -susceptible chickens; and analysis of TCR variable beta chain gene family expression suggests that MD lymphomas have polyclonal origins. Identification of the neoplastically transformed cells in MD facilitates a detailed understanding of MD pathogenesis and also improves the utility of MD as a general model for herpesvirus oncology.
Collapse
Affiliation(s)
- Shane C Burgess
- Division of Immunology and Pathology, Institute for Animal Health, Compton, United Kingdom.
| | | |
Collapse
|
22
|
Zhong MC, Kerlero de Rosbo N, Ben-Nun A. Multiantigen/multiepitope–directed immune-specific suppression of “complex autoimmune encephalomyelitis” by a novel protein product of a synthetic gene. J Clin Invest 2002. [DOI: 10.1172/jci0215692] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
23
|
Borghans JAM, De Boer RJ. Memorizing innate instructions requires a sufficiently specific adaptive immune system. Int Immunol 2002; 14:525-32. [PMID: 11978782 DOI: 10.1093/intimm/14.5.525] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
During its primary encounter with a pathogen, the immune system has to decide which type of immune response is most appropriate. Based on signals from the innate immune system and the immunological context in which the pathogen is presented, responding lymphocytes will adopt a particular phenotype, e.g. secrete a particular profile of cytokines. Once stimulated, lymphocytes store the appropriate type of response by differentiating from a naive to a memory phenotype. This allows the appropriate type of immune reaction to be regenerated upon re-stimulation of those memory clones. We developed a computer simulation model in which cross-reacting effector/memory clones contribute to the immunological context of pathogens. If a pathogen is recognized by both naive clones and pre-existing effector/memory clones, the naive lymphocytes adopt the effector mechanism of the memory clone. The adaptive immune system thereby stores immunological decisions and somatically learns to induce the right type of immune response to pathogens sharing epitopes. The influence of effector/memory lymphocytes may be detrimental when they cross-react to new pathogens that require a different kind of immune response. Here, we show that the immune system needs to be sufficiently specific to avoid such mistakes and to profit from the information that is stored in effector/memory lymphocytes. Repertoire diversity is required to reconcile this specificity with reactivity against many pathogens.
Collapse
Affiliation(s)
- José A M Borghans
- Lymphocyte Population Biology, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France.
| | | |
Collapse
|
24
|
Papassavas AC, Barnardo MCNM, Bunce M, Welsh KI. Is there MHC Class II restriction of the response to MHC Class I in transplant patients? Transplantation 2002; 73:642-51. [PMID: 11889447 DOI: 10.1097/00007890-200202270-00030] [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: 11/26/2022]
Abstract
BACKGROUND In this study, we evaluated distinct HLA-DRB1 alleles to determine class II restriction of the production of HLA-A2-specific antibodies in renal transplant patients. METHODS Data from 217 renal transplant patients who received an HLA-A2-mismatched renal graft were analyzed with regard to HLA-A2 humoral responsiveness. High-resolution DNA typing of class II HLA-DR alleles was performed by polymerase chain reaction-sequence-specific primer. Patients who had one of the following eight HLA-DRB1 alleles were included in the study: -*0101, -*0301, -*0401, -*0701, -*1101, -*1301, -*1401, and -*1501. Serum samples were screened posttransplantation with the standard complement-dependent cytotoxicity procedure. In addition, recombinant HLA-A2 monomers (the "MonoLISA" assay) were used as a target for the detection of HLA-A2 group-specific antibodies. The following HLA-A2 amino acid positions (termed "epitopes") that are responsible for the induction of an antibody response were defined: 74H, 65-66GK, 62G, 114H, 142-145TTKH, and 107W-127K. The definition of the "HLA-DR permittors" of anti-HLA-A2 response was based on a "class II restriction table" designed for this purpose. Prediction of immunogenic and/or nonimmunogenic HLA-A2 peptides was based on an MHC database. RESULTS The HLA-DRB1-*0101 and -*1401 alleles had a trend toward a positive correlation with the production of HLA class I-specific antibodies against the HLA-A2 shared (public) epitopes 65-66GK and -62G, respectively. Only the DRB1-*1501 allele had higher trend toward a positive correlation with the production of antibodies against the HLA-A2 private (74H) epitope. In 42 patients with the HLA-DRB1-*1501 allele, 11 (26%) patients produced HLA-specific antibodies against the HLA-A2 group of epitope(s). Moreover, in these patients, spreading of the alloreactivity against "other" HLA antigens was detected. Many of these other HLA antigens did not belong to HLA-A2 group but had newly defined shared epitopes with this group. Furthermore, the epitope prediction, based on an MHC database, revealed differences in the ligation strength (score) to the HLA allele (class I and II) for a specific HLA-A2 peptide in the 42 patients (responders and nonresponders). CONCLUSIONS The data presented in this paper suggest that the HLA class II allele and the type of the bound allopeptide may influence the humoral and cellular response. The immunogenicity of these allopeptides could be predicted with an MHC database (high-scored peptide=activating peptide and low-scored peptide=suppressor peptide). In the future, production of synthetic peptide analogues, on the basis of these predictions, could be used for induction of T-cell anergy and/or tolerance. In the short term, algorithms, on the basis of our approach, could be tested for influence on graft survival and allosensitization in current high-quality data sets.
Collapse
Affiliation(s)
- Andreas C Papassavas
- Transplantation Immunology, Oxford Transplant Centre, Churchill Hospital, Oxford Radcliffe Hospitals Trust, United Kingdom.
| | | | | | | |
Collapse
|
25
|
Kimball P, Elswick RK, Shiffman M. Ethnicity and cytokine production gauge response of patients with hepatitis C to interferon-alpha therapy. J Med Virol 2001. [PMID: 11596086 DOI: 10.1002/jmv.2065] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Interferon is the primary treatment for hepatitis C virus (HCV). However, the long-term success rate is low particularly for African Americans relative to Caucasians and may be due to differential immune abilities. This study compared cytokine production from PHA-stimulated peripheral blood from 25 healthy and 40 HCV-infected African Americans and Caucasians. HCV patients were designated as IFN responders or nonresponders based on outcome after therapy. Ethnicity and genotype were associated with IFN response. IFN responders were 100% Caucasian, whereas nonresponders were 67% Caucasian and 33% African American (P = 0.01). Genotype 1 was present in 100% nonresponders and 50% responders (P < 0.05). Age, sex, liver histology, ALT, and viral titers were equivalent (ns). Cytokine production from healthy individuals showed ethnic variation in cytokine levels. Healthy African Americans produced greater amounts of IL-2 (P = 0.06), TNF-alpha (P = 0.06) and less IL-10 (P = 0.05) than healthy Caucasians. In contrast, IFN-gamma and TGF-beta levels were equivalent. Pretherapy cytokine production among HCV patients showed a similar pattern of ethnic variation. African American nonresponders produced more IL-2 (P = 0.06) and TNF-alpha (P = 0.02) than Caucasian nonresponders. Cytokine levels among Caucasian and African American nonresponders were equivalent (P = ns) to ethnically matched healthy individuals whereas Caucasian responders produced subnormal levels of IL-10 (P < 0.05) and TGF-beta (P < 0.05). Since all African Americans failed IFN therapy, cytokine production could not be compared with therapeutic outcome. However, comparison of cytokine production among Caucasians showed that responders produced less IL-10 (P < 0.001) and more TGF-beta (P = 0.06) than nonresponders and predicted Caucasian nonresponders with 83% sensitivity and 96% specificity. HCV genotype was not relevant to cytokine production (P = ns). Distribution of cytokine genetic polymorphisms (TNF-alpha, TNF-beta, IL-10, TGF-beta) was equivalent in all ethnic groups and did not predict clinical nonresponders. In summary, it appears that ethnicity may contribute to variable immune responses and therapeutic outcome. The cytokine profile among African Americans suggests a more robust immune response, which may complicate therapy with IFN. In contrast, the subnormal cytokine production among Caucasian responders may be more permissive to IFN therapy. Pretherapy cytokine production may allow prediction of drug resistance among Caucasians.
Collapse
Affiliation(s)
- P Kimball
- Department of Surgery, Medical College of Virginia at the Virginia Commonwealth University, Richmond, VA 23838, USA.
| | | | | |
Collapse
|
26
|
Artik S, Haarhuis K, Wu X, Begerow J, Gleichmann E. Tolerance to nickel: oral nickel administration induces a high frequency of anergic T cells with persistent suppressor activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6794-803. [PMID: 11739495 DOI: 10.4049/jimmunol.167.12.6794] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We adapted our mouse model of allergic contact hypersensitivity to nickel for the study of tolerance. Sensitization in this model is achieved by the administration of nickel ions with H(2)O(2); nickel ions alone are unable to prime naive T cells, but can restimulate primed ones. A 4-wk course of oral or i.p. administration of 10 mM NiCl(2) to naive mice induced tolerance, preventing the induction of hypersensitivity for at least 20 wk; long term desensitization of nickel-sensitized mice, however, required continuous NiCl(2) administration. When splenic T cells of orally tolerized donors, even after a treatment-free interval of 20 wk, were transferred to naive recipients, as with lymph node cells (LNC), they specifically prevented sensitization of the recipients. The LNC of such donors were anergic, because upon in vivo sensitization with NiCl(2) in H(2)O(2) and in vitro restimulation with NiCl(2), they failed to show the enhanced proliferation and IL-2 production as seen with LNC of mice not tolerized before sensitization. As few as 10(2) bulk T cells, consisting of both CD4(+) and CD8(+) cells, were able to specifically transfer tolerance to nickel. A hypothesis is provided to account for this extraordinarily high frequency of nickel-reactive, suppressive T cells; it takes into account that nickel ions fail to act as classical haptens, but form versatile, unstable metal-protein and metal-peptide complexes. Furthermore, a powerful amplification mechanism, such as infectious tolerance, must operate which allows but a few donor T cells to tolerize the recipient.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Administration, Oral
- Adoptive Transfer
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/transplantation
- Cells, Cultured
- Clonal Anergy
- Dermatitis, Allergic Contact/drug therapy
- Dermatitis, Allergic Contact/immunology
- Dermatitis, Allergic Contact/metabolism
- Female
- Hydrogen Peroxide/pharmacology
- Injections, Intraperitoneal
- Interleukin-2/biosynthesis
- Kinetics
- Lymph Nodes/immunology
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Nickel/administration & dosage
- Nickel/pharmacokinetics
- Nickel/pharmacology
- Spleen/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/transplantation
Collapse
Affiliation(s)
- S Artik
- Division of Immunology and Allergology, Heinrich Heine University, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
27
|
van Rood JJ, Claas F. Both self and non-inherited maternal HLA antigens influence the immune response. IMMUNOLOGY TODAY 2000; 21:269-73. [PMID: 10825738 DOI: 10.1016/s0167-5699(00)01628-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Recent studies suggest that exposure of the fetus and newborn to non-inherited maternal major histocompatibility complex HLA antigens (NIMA) has a life-long effect on allograft recognition that could influence tolerance of organ grafts. NIMA also appear to influence disease susceptibility. Here, Jon van Rood and Frans Claas discuss evidence that three HLA haplotypes, those inherited from the parents plus NIMA, shape the immune response.
Collapse
Affiliation(s)
- J J van Rood
- Dept of Immunohematology and Blood Transfusion, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands.
| | | |
Collapse
|