Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Rev Med Interne ; 43(2): 89-97, 2022 Feb.
Article in French | MEDLINE | ID: mdl-35033384

ABSTRACT

Anticytoplasmic neutrophil antibodies (ANCA)-associated vasculitis (AAV) are rare systemic immune-mediated diseases characterized by small vessel necrotizing vasculitis and/or respiratory tract inflammation. Over the last 2 decades, anti-MPO vasculitis mouse model has enlightened the role of ANCA, neutrophils, complement activation, T helper cells (Th1, Th17) and microbial agents. In humans, CD4T cells have been extensively studied, while the dramatic efficacy of rituximab demonstrated the key role of B cells. Many areas of uncertainty remain, such as the driving force of GPA extra-vascular granulomatous inflammation and the relapse risk of anti-PR3 AAV pathogenesis. Animal models eventually led to identify complement activation as a promising therapeutic target. New investigation tools, which permit in depth immune profiling of human blood and tissues, may open a new era for the studying of AAV pathogenesis.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Animals , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/drug therapy , Antibodies, Antineutrophil Cytoplasmic , Disease Models, Animal , Humans , Inflammation , Mice , Neutrophils
2.
Mucosal Immunol ; 9(2): 539-49, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26329427

ABSTRACT

Crohn's disease and ulcerative colitis, the two major forms of inflammatory bowel diseases (IBDs), are characterized by high levels of IL-22 production. Rodent studies revealed that this cytokine is protective during colitis but whether this is true in IBDs is unclear. We show here that levels of the soluble inhibitor of IL-22, interleukin 22-binding protein (IL-22BP), are significantly enhanced during IBDs owing to increased numbers of IL-22BP-producing eosinophils, that we unexpectedly identify as the most abundant source of IL-22BP protein in human gut. In addition, using IL-22BP-deficient rats, we confirm that endogenous IL-22BP is effective at blocking protective actions of IL-22 during acute colitis. In conclusion, our study provides new important insights regarding the biology of IL-22 and IL-22BP in the gut and indicates that protective actions of IL-22 are likely to be suboptimal in IBDs thus making IL-22BP a new relevant therapeutic target.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Eosinophils/immunology , Interleukins/immunology , Receptors, Interleukin/immunology , Adult , Animals , Case-Control Studies , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/pathology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colon/immunology , Colon/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Dextran Sulfate , Disease Models, Animal , Eosinophils/metabolism , Eosinophils/pathology , Female , Gene Expression Regulation , Humans , Interleukins/genetics , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Signal Transduction , Interleukin-22
3.
Mucosal Immunol ; 7(1): 101-13, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23653115

ABSTRACT

Interleukin-22 (IL-22) is mainly produced at barrier surfaces by T cells and innate lymphoid cells and is crucial to maintain epithelial integrity. However, dysregulated IL-22 action leads to deleterious inflammation and is involved in diseases such as psoriasis, intestinal inflammation, and cancer. IL-22 binding protein (IL-22BP) is a soluble inhibitory IL-22 receptor and may represent a crucial regulator of IL-22. We show both in rats and mice that, in the steady state, the main source of IL-22BP is constituted by a subset of conventional dendritic cells (DCs) in lymphoid and non-lymphoid tissues. In mouse intestine, IL-22BP was specifically expressed in lamina propria CD103(+)CD11b(+) DC. In humans, IL-22BP was expressed in immature monocyte-derived DC and strongly induced by retinoic acid but dramatically reduced upon maturation. Our data suggest that a subset of immature DCs may actively participate in the regulation of IL-22 activity in the gut by producing high levels of IL-22BP.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Receptors, Interleukin/genetics , Tretinoin/pharmacology , Animals , CD4 Antigens/metabolism , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Profiling , Humans , Intestinal Mucosa/metabolism , Intestines/immunology , Male , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Organ Specificity/genetics , Protein Isoforms , Rats , Receptors, Interleukin/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism
4.
Am J Transplant ; 9(4): 697-708, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19344461

ABSTRACT

Priming of recipients by DST induces long-term survival of mismatched allografts in adult rats. Despite these recipients developing inducible T regulatory cells able to transfer long-term graft survival to a secondary host, a state of chronic rejection is also observed. We revisited the molecular donor MHC targets of the cellular response in acute rejection and analyzed the cellular and humoral responses in recipients with long-term graft survival following transplantation. We found three immunodominant peptides, all derived from LEW.1W RT1.D(u) molecules to be involved in acute rejection of grafts from unmodified LEW.1A recipients. Although the direct pathway of allorecognition was reduced in DST-treated recipients, the early CD4+ indirect pathway response to dominant peptides was almost unimpaired. We also detected early and sustained antidonor class I and II antibody subtypes with diffuse C4d deposits on graft vessels. Finally, long-term accepted grafts displayed leukocyte infiltration, endarteritis and fibrosis, which evolved toward vascular narrowing at day 100. Altogether, these data suggest that the chronic graft lesions developed in long-term graft recipients are the result of progressive humoral injury associated with a persisting indirect T helper response. These features may represent a useful model for understanding and manipulating chronic active antibody-mediated rejection in human.


Subject(s)
CD4 Antigens/immunology , Graft Rejection/immunology , Graft Survival/immunology , Isoantibodies/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Antibody Formation , Blood Transfusion , Histocompatibility Antigens/genetics , Humans , Immunity, Cellular , Polymorphism, Genetic , Rats , Rats, Inbred BN , Rats, Inbred Lew , T-Lymphocytes, Helper-Inducer/immunology , Tissue Donors
5.
Am J Transplant ; 7(11): 2472-82, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17868070

ABSTRACT

Regulatory T cells have been described to specifically accumulate at the site of regulation together with effector T cells and antigen-presenting cells, establishing a state of local immune privilege. However the mechanisms of this interplay remain to be defined. We previously demonstrated, in a fully MHC mismatched rat cardiac allograft combination, that a short-term treatment with a deoxyspergualine analogue, LF15-0195, induces long-term allograft tolerance with a specific expansion of regulatory CD4+CD25+T cells that accumulate within the graft. In this study, we show that following transfer of regulatory CD4+T cells to a secondary irradiated recipient, regulatory CD25+Foxp3+ and CD25+Foxp3(-) CD4+T cells accumulate at the graft site and induce graft endothelial cell expression of Indoleamine 2, 3-dioxygenase (IDO) by an IFNgamma-dependent mechanism. Moreover, in vivo transfer of tolerance can be abrogated by blocking IFNgamma or IDO, and anti-IFNgamma reduces the survival/expansion of alloantigen-induced regulatory Foxp3+CD4+T cells. Together, our results demonstrate interrelated mechanisms between regulatory CD4+CD25+T cells and the graft endothelial cells in this local immune privilege, and a key role for IFNgamma and IDO in this process.


Subject(s)
CD4 Antigens/immunology , Heart Transplantation/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interferon-gamma/physiology , Interleukin-2 Receptor alpha Subunit/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Transplantation, Homologous/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Coculture Techniques , Endothelium, Vascular/cytology , Enzyme Induction , Forkhead Transcription Factors/physiology , Guanidines/pharmacology , Heart Transplantation/pathology , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Lymphocyte Culture Test, Mixed , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology
6.
Proc Natl Acad Sci U S A ; 98(21): 12162-7, 2001 Oct 09.
Article in English | MEDLINE | ID: mdl-11593031

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is well recognized for its role in mediating innate immune responses. However, the mechanisms of TNF-alpha that influence the adaptive immune response to virus infections are not well understood. In this study, we have investigated the role of TNF-alpha in activating the cellular and humoral responses to systemic viral challenge with recombinant replication-defective adenovirus (rAd). Evaluation of T cell function in TNF-alpha-deficient (TNFKO) mice revealed impaired virus-specific proliferation of T cells derived from the draining lymph nodes of the liver. Analysis of dendritic cells (DC) isolated from local draining lymph nodes after systemic challenge showed that DC from TNFKO mice were relatively immature compared with those from strain-matched wild-type mice. In vitro, TNF-alpha was required to mature DC efficiently during virus-mediated stimulation. Adoptive transfer of primed, mature DC into TNFKO mice restored T cell responses and reconstituted anti-adenovirus antibody responses. Thus, TNF-alpha plays a significant role in the maturation of DC after adenovirus challenge both in vitro and in vivo, highlighting the importance of this innate cytokine in activating adaptive immunity to viral challenge.


Subject(s)
Adenoviridae Infections/immunology , Dendritic Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Adaptation, Physiological/immunology , Adenoviruses, Human/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Cell Differentiation , Defective Viruses/immunology , Dendritic Cells/cytology , Immunity, Active , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/genetics
7.
Curr Opin Nephrol Hypertens ; 10(3): 349-54, 2001 May.
Article in English | MEDLINE | ID: mdl-11342796

ABSTRACT

The present review briefly addresses the most recent knowledge acquired in the field of transplant immunology. A particular emphasis is placed on articles published during the past 12-18 months that have focused on allorecognition, dendritic cells and tolerance.


Subject(s)
Organ Transplantation , Transplantation Immunology , Animals , Dendritic Cells/immunology , Graft Rejection , Humans , Mice , Rats , Transplantation, Homologous
8.
J Exp Med ; 192(10): 1467-78, 2000 Nov 20.
Article in English | MEDLINE | ID: mdl-11085748

ABSTRACT

Proper lymph node (LN) development requires tumor necrosis factor-related activation-induced cytokine (TRANCE) expression. Here we demonstrate that the defective LN development in TRANCE(-/)- mice correlates with a significant reduction in lymphotoxin (LT)alphabeta(+)alpha(4)beta(7)(+)CD45(+)CD4(+)CD3(-) cells and their failure to form clusters in rudimentary mesenteric LNs. Transgenic TRANCE overexpression in TRANCE(-/)- mice results in selective restoration of this cell population into clusters, and results in full LN development. Transgenic TRANCE-mediated restoration of LN development requires LTalphabeta expression on CD45(+) CD4(+)CD3(-) cells, as LNs could not be induced in LTalpha(-/)- mice. LTalpha(-/)- mice also showed defects in the fate of CD45(+)CD4(+)CD3(-) cells similar to TRANCE(-/)- mice. Thus, we propose that both TRANCE and LTalphabeta regulate the colonization and cluster formation by CD45(+) CD4(+)CD3(-) cells in developing LNs, the degree of which appears to correlate with the state of LN organogenesis.


Subject(s)
Carrier Proteins/metabolism , Lymph Nodes/growth & development , Membrane Glycoproteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , B-Lymphocytes , CD3 Complex , CD4 Antigens , Leukocyte Common Antigens , Mice , Mice, Transgenic , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Spleen
9.
J Immunol ; 165(8): 4202-8, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035052

ABSTRACT

Dendritic cells (DCs) are a rare population of leukocytes specialized in Ag processing and presentation to T cells. We have previously shown that cultured rat splenic DCs exhibit a cytotoxic activity against selected target cells. In this study, we analyzed this function in DCs freshly prepared from lymphoid organs using the DC-specific OX62 mAb and magnetic beads. Freshly extracted splenic DCs, but not lymph node and thymic DCs, exhibited a strong and moderate cytotoxic activity against YAC-1 and K562 target cells, respectively. FACS analyses showed that spleen contained a minor subset (10-15%) of CD4(+) and class II(int) DCs that also expressed the OX41 Ag and the lymphoid-related Ags CD5 and CD90 (Thy-1) and a major (80-85%) subset of CD4(-)/OX41(-)/CD5(-) and class II(int) DCs. The cytotoxic activity of splenic DCs was strictly restricted to the CD4(-) DCs, a subset poorly represented in LN and thymus. Contrasting with our previous report using cultured splenic DCs, freshly isolated splenic DCs killed YAC-1 cells using a Ca(2+)-independent mechanism, but this function did not appear mediated by Fas ligand, TNF-related apoptosis-inducing ligand, or TNF-alpha. Therefore, rat DCs contain a subset of naturally cytolytic cells that could play a role in both innate and acquired immune responses. Together with our previous report, these data suggest that rat DCs can use two mechanisms of cytotoxicity depending on their maturation/activation state.


Subject(s)
Cytotoxicity, Immunologic , Dendritic Cells/immunology , Animals , Apoptosis/immunology , Apoptosis Regulatory Proteins , CD4 Antigens/biosynthesis , Cell Line , Cell Separation , Dendritic Cells/metabolism , Fas Ligand Protein , Immunophenotyping , Killer Cells, Natural/immunology , Leukemia L5178 , Ligands , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Membrane Glycoproteins/physiology , Mice , Organ Specificity/immunology , Perforin , Pore Forming Cytotoxic Proteins , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Spleen/cytology , Spleen/immunology , Spleen/metabolism , TNF-Related Apoptosis-Inducing Ligand , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology
10.
J Immunol ; 165(1): 96-101, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861040

ABSTRACT

Donor-specific allograft tolerance can be induced in the adult rat by pregraft donor-specific blood transfusion (DST). This tolerance appeared to be mediated by regulatory cells and to the production of the suppressive cytokine TGF-beta1. A potential immunoregulatory CD8+ clone bearing a Vbeta18-Dbeta1-Jbeta2.7 TCR gene rearrangement was previously identified in DST-treated recipients. To assess the functional role of this T cell clone in the induction of tolerance by DST, we have vaccinated DST-treated recipients with a plasmid construct encoding for the Vbeta18-Dbeta1-Jbeta2.7 TCR beta-chain. DST-induced allograft tolerance was abolished by anti-TCR Vbeta18-Dbeta1-Jbeta2.7 DNA vaccination in six of seven recipients, whereas vaccination with the vector alone, or with the construct encoding a TCR Vbeta13 beta-chain, had no effect. However, the transcript number of the Vbeta18-Dbeta1-Jbeta2.7 chain was unchanged in allografts from vaccinated DST-treated rats, suggesting that this clone was not depleted by vaccination, but rather was altered in its function. Moreover, TCR Vbeta18-Dbeta1-Jbeta2.7 DNA vaccination restored the anti-donor alloantibody production, partially restore the capacity of spleen cells from tolerized recipients to proliferate in vitro against donor cells, and decreased the inhibitory effect of TGF-beta1, seen in DST-treated recipients, in spleen cells from vaccinated DST-treated ones. This study strongly suggests that this CD8+ TCR Vbeta18-Dbeta1-Jbeta2.7 T cell clone has an effective immunoregulatory function in allograft tolerance induced by DST.


Subject(s)
Blood Transfusion , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Genes, T-Cell Receptor beta/immunology , Heart Transplantation/immunology , Immune Tolerance , Vaccines, DNA/immunology , Animals , Clonal Deletion/genetics , Clone Cells , Cytokines/biosynthesis , Cytokines/genetics , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Epitopes, T-Lymphocyte/genetics , Graft Rejection/genetics , Graft Rejection/immunology , Immune Tolerance/genetics , Immunoglobulin G/biosynthesis , Injections, Intramuscular , Isoantibodies/biosynthesis , Male , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Lew , Spleen/cytology , Spleen/immunology , Transplantation, Heterotopic , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
11.
Inflammation ; 24(4): 347-56, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10850856

ABSTRACT

The effect of high oxygen concentrations on lungs of neonatal rats was studied. In addition, some oxygen-exposed animals were treated with either dexamethasone or thalidomide. No gross histologic changes were noted in the lungs following exposure to 95% oxygen nor were there changes in the total number or the phenotypic distribution of BAL cells obtained from these lungs compared to lungs from air exposed (control) neonatal rats. The majority of the BAL cells were CD45+ leukocytes (macrophages). However, when BAL cells were exposed to LPS in vitro, TNF-alpha production was higher in cells from rats exposed to 95% oxygen compared to cells from rats exposed to ambient air. In addition, lung TNF-alpha and IL-6 mRNA levels were increased after exposure to 95% oxygen. In the lungs of animals treated with either dexamethasone or thalidomide, TNF-alpha mRNA levels were reduced, while only dexamethasone treatment also reduced IL-6 mRNA levels.


Subject(s)
Dexamethasone/pharmacology , Hyperoxia/drug therapy , Hyperoxia/genetics , Lung/drug effects , Lung/metabolism , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/genetics , Animals , Animals, Newborn , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Female , Gene Expression/drug effects , Hyperoxia/metabolism , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Interleukin-6/genetics , Lung Injury , Macrophages, Alveolar/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
12.
J Exp Med ; 191(3): 495-502, 2000 Feb 07.
Article in English | MEDLINE | ID: mdl-10662795

ABSTRACT

Mature dendritic cells (DCs) are powerful antigen presenting cells that have the unique capacity to migrate to the T cell zone of draining lymph nodes after subcutaneous injection. Here we report that treatment of antigen-pulsed mature DCs with tumor necrosis factor (TNF)-related activation-induced cytokine (TRANCE), a TNF family member, before immunization enhances their adjuvant capacity and elicits improved T cell priming in vivo, such that both primary and memory T cell immune responses are enhanced. By enumerating migratory DCs in the draining lymph nodes and by studying their function in stimulating naive T cells, we show that one of the underlying mechanisms for enhanced T cell responses is an increase in the number of ex vivo antigen-pulsed DCs that are found in the T cell areas of lymph nodes. These results suggest that the longevity and abundance of mature DCs at the site of T cell priming influence the strength of the DC-initiated T cell immunity in situ. Our findings have the potential to improve DC-based immunotherapy; i.e., the active immunization of humans with autologous DCs that have been pulsed with clinically significant antigens ex vivo.


Subject(s)
Carrier Proteins/pharmacology , Dendritic Cells/drug effects , Lymph Nodes/immunology , Membrane Glycoproteins/pharmacology , Adjuvants, Immunologic , Animals , CD40 Ligand , Carrier Proteins/genetics , Cell Count/drug effects , Cells, Cultured , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Fluoresceins , Fluorescent Dyes , Granulocyte-Macrophage Colony-Stimulating Factor , Immunization , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , Tuberculin , Tumor Necrosis Factor-alpha/genetics
13.
J Pathol ; 189(3): 378-86, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10547600

ABSTRACT

CD95 ligand expression has been observed in various malignancies. Studying the CD95 ligand (CD95L) and receptor (CD95) system in eight non-malignant mammary tissues and 40 breast cancer tissues, mRNA and protein expression was determined by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) and immunofluorescence. mRNA levels of CD95L correlated positively (r=0.90; p< 0.01) and transmembrane CD95 inversely (r=-0.88; p< 0.01) with histopathological grading of the breast tumours: CD95L mRNA levels were low in adenomas, but increased by 20-fold in grade I, 120-fold in grade II, and 310-fold in grade III breast cancer. In contrast, CD95 mRNA levels were low in high-grade carcinomas, but high in benign mammary tissues. Since CD95L acts as an efficient inducer of apoptosis in CD95(+) cells, apoptotic cells were identified on the tissue sections. Tumour-infiltrating lymphocytes and stromal cells in close proximity to CD95L-expressing breast cancer underwent apoptosis. As a functional test, CD95(+) target cells were cultured on breast cancer tissue sections. The target cells underwent apoptosis when cultured on breast cancer sections, but could be rescued when CD95L was specifically blocked by a CD95-Fc fusion molecule. The data suggest an inverse regulation of CD95 ligand and receptor expression during dedifferentiation of breast cancer. Killing of bystander cells by the CD95L-expressing breast tumour could be involved in tissue invasion.


Subject(s)
Antigens, Neoplasm/metabolism , Breast Neoplasms/immunology , Membrane Glycoproteins/metabolism , fas Receptor/metabolism , Adult , Aged , Apoptosis , Breast Neoplasms/pathology , Cell Differentiation , Fas Ligand Protein , Female , Fluorescent Antibody Technique , Humans , Jurkat Cells/metabolism , Ligands , Membrane Glycoproteins/genetics , Middle Aged , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction
14.
Transplantation ; 68(7): 913-21, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10532525

ABSTRACT

Lymphocytes express antigen receptors that allow the immune system to specifically recognize antigens. In transplantation, T cells play a critical role in the rejection process, and different protocols inhibiting T cell-mediated alloreactivity efficiently achieve prolongation of allograft survival. T cells can interact with alloantigens by two ways, either by the "indirect" pathway that correspond to the physiological mechanism of T cell immune recognition, or through the "direct" pathway where they recognize alloantigens directly on the surface of donor cells. If some T cells are specifically activated in allorecognition, one should be able to indirectly detect this "selection" by analyzing the T cell receptor usage that could be biased and reflect the preferential amplification of alloreactive lymphocyte subsets. Nevertheless compared with disease states such as cancer or autoimmunity the T cell receptor repertoire is still largely uncharacterized. We review the current results available on T cell repertoire usage in transplantation studies involving humans or various animal models. The T cell receptor repertoire involved in transplantation (restricted or unrestricted) and the features potentially common to alloimmune responses will be discussed.


Subject(s)
Isoantigens/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Transplantation, Homologous/immunology , Animals , Humans
15.
Eur J Immunol ; 29(6): 1919-24, 1999 06.
Article in English | MEDLINE | ID: mdl-10382754

ABSTRACT

Donor-specific allograft tolerance can be induced in adult rats by pregraft donor-specific blood transfusion (DST). We have previously shown that DST elicits in recipients the expansion of a donor-specific CD8+ T cell clone displaying the Vbeta18-Dbeta1-Jbeta2.7 TCR rearrangement, which rapidly infiltrates allografts after transplantation, suggesting a regulatory function for this clone in DST-induced tolerance. In this study, recipients were pretreated before grafting, using an anti-CD8 monoclonal antibody to deplete CD8+ T cells. CD8 depletion before DST and transplantation abrogated allograft tolerance, and the CD8+ T cell clone was absent from allografts. These effects were not observed when CD8 depletion was performed after DST but before transplantation. We conclude that CD8+ T cells play a role in the induction of allograft tolerance by DST.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Graft Survival/immunology , Acute Disease , Animals , Antibodies, Monoclonal/administration & dosage , Antilymphocyte Serum/administration & dosage , Base Sequence , Blood Transfusion , DNA Probes/genetics , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Heart Transplantation/immunology , Immune Tolerance/genetics , Lymphocyte Depletion , Male , Rats , Rats, Inbred Lew , Tissue Donors , Transplantation, Homologous
16.
J Leukoc Biol ; 65(6): 715-24, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10380891

ABSTRACT

Tumor necrosis factor (TNF)-related activation-induced cytokine (TRANCE) is a new member of the TNF family emerging as a key regulator of the immune system and of bone development and homeostasis. TRANCE is expressed on activated T cells and activates mature dendritic cells (DC), suggesting that it plays a role in the T cell-DC interaction during an immune response. Furthermore, TRANCE is expressed on osteoblasts stimulated with vitamin D3, dexamethasone, and parathyroid hormone. TRANCE, when expressed on osteoblasts, induces osteoclastogenesis and osteoclast activation, suggesting that it links known calciotropic hormones to bone resorption. TRANCE mediates its effects via the TRANCE-receptor (TRANCE-R/RANK), whereas its activity can be inhibited by the soluble decoy receptor osteoprotegerin/osteoclast inhibitory factor (OPG/OCIF). OPG can be neutralized by another TNF-family member, the TNF-related apoptosis-inducing ligand (TRAIL), suggesting that TRANCE is part of a complex cytokine network that regulates a diverse set of functions. We will discuss the current literature describing TRANCE and its receptors and its role in controlling DC and osteoclast function.


Subject(s)
Carrier Proteins/physiology , Dendritic Cells/physiology , Membrane Glycoproteins/physiology , Osteoclasts/physiology , Animals , Humans , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology
17.
J Biol Chem ; 274(19): 13613-8, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224132

ABSTRACT

Tumor necrosis factor (TNF)-related activation-induced cytokine (TRANCE), a member of the TNF family, is a dendritic cell survival factor and is essential for osteoclastogenesis and osteoclast activation. In this report we demonstrate (i) that TRANCE, like TNF-alpha, is made as a membrane-anchored precursor, which is released from the plasma membrane by a metalloprotease; (ii) that soluble TRANCE has potent dendritic cell survival and osteoclastogenic activity; (iii) that the metalloprotease-disintegrin TNF-alpha convertase (TACE) can cleave immunoprecipitated TRANCE in vitro in a fashion that mimics the cleavage observed in tissue culture cells; and (iv) that in vitro cleavage of a TRANCE ectodomain/CD8 fusion protein and of a peptide corresponding to the TRANCE cleavage site by TACE occurs at the same site that is used when TRANCE is shed from cells into the supernatant. We propose that the TRANCE ectodomain is released from cells by TACE or a related metalloprotease-disintegrin, and that this release is an important component of the function of TRANCE in bone and immune homeostasis.


Subject(s)
Carrier Proteins/metabolism , Dendritic Cells/cytology , Membrane Glycoproteins/metabolism , Metalloendopeptidases/metabolism , Osteoclasts/cytology , ADAM Proteins , ADAM17 Protein , Amino Acid Sequence , Animals , COS Cells , Cell Differentiation , Cell Line , Cell Survival , Humans , Hydrolysis , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B
18.
J Exp Med ; 189(7): 1025-31, 1999 Apr 05.
Article in English | MEDLINE | ID: mdl-10190893

ABSTRACT

CD40 ligand (CD40L), a tumor necrosis factor (TNF) family member, plays a critical role in antigen-specific T cell responses in vivo. CD40L expressed on activated CD4(+) T cells stimulates antigen-presenting cells such as dendritic cells, resulting in the upregulation of costimulatory molecules and the production of various inflammatory cytokines required for CD4(+) T cell priming in vivo. However, CD40L- or CD40-deficient mice challenged with viruses mount protective CD4(+) T cell responses that produce normal levels of interferon gamma, suggesting a CD40L/CD40-independent mechanism of CD4(+) T cell priming that to date has not been elucidated. Here we show that CD4(+) T cell responses to viral infection were greatly diminished in CD40-deficient mice by administration of a soluble form of TNF-related activation-induced cytokine receptor (TRANCE-R) to inhibit the function of another TNF family member, TRANCE. Thus, the TRANCE/TRANCE-R interaction provides costimulation required for efficient CD4(+) T cell priming during viral infection in the absence of CD40L/CD40. These results also indicate that not even the potent inflammatory microenvironment induced by viral infections is sufficient to elicit efficient CD4(+) T cell priming without proper costimulation provided by the TNF family (CD40L or TRANCE). Moreover, the data suggest that TRANCE/TRANCE-R may be a novel and important target for immune intervention.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Carrier Proteins/physiology , Dendritic Cells/immunology , Lymphocyte Activation/physiology , Membrane Glycoproteins/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Germinal Center/immunology , Immunity, Cellular , Immunoglobulin Isotypes/immunology , Interferon-gamma/biosynthesis , Lymphocytic Choriomeningitis/immunology , Lymphokines/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Multigene Family , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Sequence Homology, Amino Acid , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology
19.
J Immunol ; 162(5): 2562-8, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10072496

ABSTRACT

TNF-related activation-induced cytokine (TRANCE) is a member of the TNF family recently identified in activated T cells. We report here that TRANCE mRNA is constitutively expressed in memory, but not naive, T cells and in single-positive thymocytes. Upon TCR/CD3 stimulation, TRANCE mRNA and surface protein expression are rapidly up-regulated in CD4+ and CD8+ T cells, which can be further enhanced on CD4+ T cells by CD28-mediated costimulation. However, TRANCE induction is significantly suppressed when cells are stimulated in the presence of IL-4, but is not modified in the presence of IFN-alpha, IFN-gamma, TGF-beta, TNF-alpha, or IL-2. High levels of TRANCE receptor expression are found on mature dendritic cells (DCs). In this study we show that activated T and B cells also express TRANCE receptor, but only at low levels. TRANCE, however, does not exert any significant effect on the proliferation, activation, or survival of those cells. In DCs, TRANCE induces the expression of proinflammatory cytokines (IL-6, IL-1) and T cell growth and differentiation factors (IL-12, IL-15) in addition to enhancing DC survival. Moreover, TRANCE cooperates with CD40 ligand or TNF-alpha to further increase the viability of DCs, suggesting that several TNF-related molecules on activated T cells may cooperatively regulate the function and survival of DCs to enhance T cell-mediated immune responses.


Subject(s)
Carrier Proteins , Cytokines/biosynthesis , Dendritic Cells/physiology , Membrane Glycoproteins/physiology , T-Lymphocyte Subsets/immunology , Animals , CD40 Ligand , Interleukin-15/physiology , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RANK Ligand , RNA, Messenger/analysis , Receptor Activator of Nuclear Factor-kappa B , Tumor Necrosis Factor-alpha/physiology
20.
Eur J Immunol ; 29(1): 317-26, 1999 01.
Article in English | MEDLINE | ID: mdl-9933114

ABSTRACT

Donor-specific tolerance to heart allograft was induced in adult Lewis rats by pregraft donor-specific blood transfusion (DST). We previously showed that this tolerant state is characterized by a dramatic inhibition of T cell and macrophage activation. In addition, tolerant animals could not mount an efficient anti-donor humoral response whereas transfer of sera from rejecting animals triggered rejection in tolerant animals. This tolerance can be abrogated by daily post-graft administration of recombinant IFN-gamma (rIFN-gamma). To elucidate the mechanisms of action of rIFN-gamma, T cell, macrophage and B cell functions were assessed in allograft recipients. IFN-gamma did not restore the expression of Th1-related cytokine mRNA or the activated macrophage product inducible nitric oxide synthase in allografts. Importantly, rIFN-gamma treatment promptly restored the anti-donor humoral response in DST-treated recipients. We conclude that rIFN-gamma treatment in DST-treated allograft recipients cannot reverse the unresponsive state of Th1 cells and macrophages infiltrating the graft, but can provide B cell help for IgG alloantibody production which is lacking in these animals.


Subject(s)
Immune Tolerance/drug effects , Interferon-gamma/pharmacology , Isoantibodies/biosynthesis , Transplantation Immunology/drug effects , Animals , Base Sequence , Blood Transfusion , Cytokines/genetics , DNA Primers/genetics , Heart Transplantation/immunology , Immunoglobulin G/biosynthesis , In Vitro Techniques , Lymphocyte Activation , Macrophage Activation , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Recombinant Proteins , Th1 Cells/immunology , Tissue Donors , Transforming Growth Factor beta/genetics , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL