Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
J Immunol ; 187(3): 1347-57, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21715686

ABSTRACT

Identification of correlates of protection for infectious diseases including malaria is a major challenge and has become one of the main obstacles in developing effective vaccines. We investigated protection against liver-stage malaria conferred by vaccination with adenoviral (Ad) and modified vaccinia Ankara (MVA) vectors expressing pre-erythrocytic malaria Ags. By classifying CD8(+) T cells into effector, effector memory (T(EM)), and central memory subsets using CD62L and CD127 markers, we found striking differences in T cell memory generation. Although MVA induced accelerated central memory T cell generation, which could be efficiently boosted by subsequent Ad administration, it failed to protect against malaria. In contrast, Ad vectors, which permit persistent Ag delivery, elicit a prolonged effector T cell and T(EM) response that requires long intervals for an efficient boost. A preferential T(EM) phenotype was maintained in liver, blood, and spleen after Ad/MVA prime-boost regimens, and animals were protected against malaria sporozoite challenge. Blood CD8(+) T(EM) cells correlated with protection against malaria liver-stage infection, assessed by estimation of number of parasites emerging from the liver into the blood. The protective ability of Ag-specific T(EM) cells was confirmed by transfer experiments into naive recipient mice. Thus, we identify persistent CD8 T(EM) populations as essential for vaccine-induced pre-erythrocytic protection against malaria, a finding that has important implications for vaccine design.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Immunologic Memory , Liver Diseases, Parasitic/immunology , Liver Diseases, Parasitic/prevention & control , Malaria/immunology , Malaria/prevention & control , Adenoviridae/genetics , Adenoviridae/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Genetic Vectors/therapeutic use , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , H-2 Antigens/immunology , Humans , Immunologic Memory/genetics , Liver Diseases, Parasitic/pathology , Malaria/pathology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Transgenes/immunology , Vaccinia/genetics , Vaccinia/immunology
2.
J Immunol ; 187(2): 835-41, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21690324

ABSTRACT

The immune system has evolved to use sophisticated mechanisms to recruit lymphocytes to sites of pathogen exposure. Trafficking pathways are precise. For example, lymphocytes that are primed by gut pathogens can, in some cases, be imprinted with CCR9 membrane receptors, which can influence migration to the small intestine. Currently, little is known about T cell trafficking to the upper respiratory tract or the relationship between effectors that migrate to the diffuse nasal-associated lymphoid tissue (d-NALT), the lower airways, and the lung. To determine whether a T cell primed by Ag from a respiratory pathogen is imprinted for exclusive trafficking to the upper or lower respiratory tract or whether descendents from that cell have the capacity to migrate to both sites, we inoculated mice by the intranasal route with Sendai virus and conducted single-cell-sequencing analyses of CD8(+) T lymphocytes responsive to a K(b)-restricted immunodominant peptide, FAPGNYPAL (Tet(+)). Cells from the d-NALT, lung airways (bronchoalveolar lavage), lung, and mediastinal lymph node were examined 10 d postinfection to determine TCR usage and clonal relationships. We discovered that 1) Tet(+) cells were heterogeneous but preferentially used TCR elements TRAV6, TRAV16, and TRBD1; 2) both N and C termini of Vα and Vß TCR junctions frequently encompassed charged residues, perhaps facilitating TCR αß pairing and interactions with a neutral target peptide; and 3) T cells in the d-NALT were often clonally related to cells in the lower respiratory tract.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Lung/immunology , Lymphoid Tissue/immunology , Nasal Mucosa/immunology , Respiratory Tract Infections/immunology , Respirovirus Infections/immunology , Sendai virus/immunology , Administration, Intranasal , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Clone Cells , Disease Models, Animal , Epitopes, T-Lymphocyte/metabolism , Female , H-2 Antigens/administration & dosage , H-2 Antigens/immunology , H-2 Antigens/metabolism , Immunodominant Epitopes/metabolism , Immunoglobulin Variable Region/metabolism , Lung/pathology , Lung/virology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nasal Mucosa/pathology , Nasal Mucosa/virology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Respirovirus Infections/pathology , Respirovirus Infections/virology , Sendai virus/isolation & purification
3.
J Immunol ; 186(2): 856-68, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21149609

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is considered an autoimmune-like disease mediated by donor CD4(+) T cells, but the origin of the autoreactive T cells is still controversial. In this article, we report that the transplantation of DBA/2 donor spleen cells into thymectomized MHC-matched allogeneic BALB/c recipients induced autoimmune-like cGVHD, although not in control syngeneic DBA/2 recipients. The donor-type CD4(+) T cells from the former but not the latter recipients induced autoimmune-like manifestations in secondary allogeneic BALB/c as well as syngeneic DBA/2 recipients. Transfer of donor-type CD4(+) T cells from secondary DBA/2 recipients with disease into syngeneic donor-type or allogeneic host-type tertiary recipients propagated autoimmune-like manifestations in both. Furthermore, TCR spectratyping revealed that the clonal expansion of the autoreactive CD4(+) T cells in cGVHD recipients was initiated by an alloimmune response. Finally, hybridoma CD4(+) T clones derived from DBA/2 recipients with disease proliferated similarly in response to stimulation by syngeneic donor-type or allogeneic host-type dendritic cells. These results demonstrate that the autoimmune-like manifestations in cGVHD can be mediated by a population of donor CD4(+) T cells in transplants that simultaneously recognize Ags presented by both donor and host APCs.


Subject(s)
Autoantigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Graft vs Host Disease/immunology , H-2 Antigens/immunology , Histocompatibility Antigens Class II/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Autoantibodies/biosynthesis , Autoantigens/administration & dosage , Autoantigens/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Chronic Disease , Clone Cells , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , Histocompatibility Antigen H-2D , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/genetics , Histocompatibility Testing , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Models, Animal , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/transplantation
4.
J Immunol ; 185(5): 2895-902, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20675595

ABSTRACT

T-bet (Tbx21), a T-box transcription factor, has been previously identified as a master regulator of type 1 T cell polarization. We have also recently shown that the genetic engineering of human dendritic cells (DCs) to express human T-bet cDNA yields type 1-polarizing APCs in vitro (1). In the present study, murine CD11c(+) DCs were transduced with a recombinant adenovirus encoding full-length murine T-bets (DC.mTbets) and analyzed for their immunomodulatory functions in vitro and in vivo. Within the range of markers analyzed, DC.mTbets exhibited a control DC phenotype and were indistinguishable from control DCs in their ability to promote allogenic T cell proliferation in MLR in vitro. However, DC.mTbets were superior to control DCs in promoting Th1 and Tc1 responses in vitro via a mechanism requiring DC-T cell interaction or the close proximity of these two cell types and that can only partially be explained by the action of DC-elaborated IL-12p70. When injected into day 7 s.c. CMS4 sarcoma lesions growing in syngenic BALB/c mice, DC.mTbets dramatically slowed tumor progression (versus control DCs) and extended overall survival via a mechanism dependent on both CD4(+) and CD8(+) T cells and, to a lesser extent, asialoGM1(+) NK cells. DC.mTbet-based therapy also promoted superior tumor-specific Tc1 responses in the spleens and tumor-draining lymph nodes of treated animals, and within the tumor microenvironment it inhibited the accumulation of CD11b(+)Gr1(+) myeloid-derived suppressor cells and normalized CD31(+) vascular structures. These findings support the potential translational utility of DC.Tbets as a therapeutic modality in the cancer setting.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Genetic Engineering/methods , Immunity, Innate/genetics , Injections, Intralesional/methods , Sarcoma, Experimental/immunology , Sarcoma, Experimental/therapy , T-Box Domain Proteins/administration & dosage , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Female , Gene Expression Regulation/immunology , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , Humans , Immunophenotyping , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Sarcoma, Experimental/mortality , Sarcoma, Experimental/virology , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Transduction, Genetic
5.
J Immunol ; 184(8): 4196-204, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20220085

ABSTRACT

There is compelling evidence that self-reactive CD8(+) T cells are a major factor in development and progression of type 1 diabetes in animals and humans. Hence, great effort has been expended to define the specificity of autoimmune CD8(+) T cells and to alter their responses. Much work has focused on tolerization of T cells using proteins or peptides. A weakness in this approach is that residual autoreactive T cells may be activated and exacerbate disease. In this report, we use a novel approach, toxin-coupled MHC class I tetramers. Used for some time to identify Ag-specific cells, in this study, we use that same property to delete the Ag-specific cells. We show that saporin-coupled tetramers can delete islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-reactive T cells in vitro and in vivo. Sequence analysis of TCRbeta-chains of IGRP(+) cells reveals the repertoire complexity in the islets is markedly decreased as NOD mice age and significantly altered in toxic tetramer-treated NOD mice. Further tetramer(+) T cells in the islets are almost completely deleted, and, surprisingly, loss of tetramer(+) T cells in the islets is long lasting. Finally, we show deletion at 8 wk of age of IGRP(+) CD8(+) T cells, but not dystophia myotonica kinase- or insulin B-reactive cells, significantly delays diabetes in NOD mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Diabetes Mellitus, Type 1/prevention & control , H-2 Antigens/administration & dosage , Immunotoxins/administration & dosage , Ribosome Inactivating Proteins, Type 1/toxicity , beta 2-Microglobulin/administration & dosage , Animals , Autoantigens/immunology , Autoantigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Death/immunology , Cell Movement/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Progression , Epitopes, T-Lymphocyte/immunology , Female , Glucose-6-Phosphatase/administration & dosage , Glucose-6-Phosphatase/biosynthesis , Glucose-6-Phosphatase/immunology , H-2 Antigens/toxicity , Histocompatibility Antigen H-2D , Immunotoxins/toxicity , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Molecular Mimicry/immunology , Proteins/administration & dosage , Proteins/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Ribosome Inactivating Proteins, Type 1/administration & dosage , Saporins , beta 2-Microglobulin/toxicity
6.
J Immunol ; 183(4): 2425-34, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19620307

ABSTRACT

Prime-boost immunization with gene-based vectors has been developed to generate more effective vaccines for AIDS, malaria, and tuberculosis. Although these vectors elicit potent T cell responses, the mechanisms by which they stimulate immunity are not well understood. In this study, we show that immunization by a single gene product, HIV-1 envelope, with alternative vector combinations elicits CD8(+) cells with different fine specificities and kinetics of mobilization. Vaccine-induced CD8(+) T cells recognized overlapping third V region loop peptides. Unexpectedly, two anchor variants bound H-2D(d) better than the native sequences, and clones with distinct specificities were elicited by alternative vectors. X-ray crystallography revealed major differences in solvent exposure of MHC-bound peptide epitopes, suggesting that processed HIV-1 envelope gave rise to MHC-I/peptide conformations recognized by distinct CD8(+) T cell populations. These findings suggest that different gene-based vectors generate peptides with alternative conformations within MHC-I that elicit distinct T cell responses after vaccination.


Subject(s)
AIDS Vaccines/genetics , AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , H-2 Antigens/genetics , H-2 Antigens/immunology , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Line , Cells, Cultured , Clone Cells , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Genetic Vectors/metabolism , H-2 Antigens/administration & dosage , H-2 Antigens/metabolism , HIV Envelope Protein gp120/administration & dosage , HIV Envelope Protein gp120/metabolism , Histocompatibility Antigen H-2D , Immunization, Secondary , Mice , Mice, Inbred BALB C , Protein Binding/genetics , Protein Binding/immunology
7.
J Immunol ; 183(1): 181-90, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542429

ABSTRACT

Suppressing the abnormalities associated with asthma has been difficult to accomplish using immunotherapy or vaccination once the disease is established. The effector cells necessary for effective immunization/vaccination and immunotherapy of asthma are also not well understood. Therefore, we vaccinated allergen (OVA)-sensitized mice to determine whether therapeutic immunization could suppress airway hyperresponsiveness (AHR) and inflammation and to identify key immune effector cells and cytokines. Mice were immunized with a vaccine comprised of Ag and cationic liposome-DNA complexes (CLDC), a vaccine which has previously been shown to elicit strong CD4(+) and CD8(+) T cell responses and activation of Th1 immunity. We showed that immunization with the OVA-CLDC vaccine significantly suppressed AHR, eosinophilia, goblet cell metaplasia, and Th2 cytokine production. In contrast, immunization with CLDC alone suppressed eosinophilia and Th2 cytokine production, but failed to suppress AHR and goblet cell changes. Using adoptive transfer experiments, we found that suppression of AHR was mediated by Ag-specific CD8(+) T cells and was dependent on IFN-gamma production by the transferred T cells. Thus, we conclude that generation of strong, allergen-specific CD8(+) T cell responses by immunization may be capable of suppressing AHR and allergic airway inflammation, even in previously sensitized and challenged mice.


Subject(s)
Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/prevention & control , CD8-Positive T-Lymphocytes/immunology , Immunosuppressive Agents/administration & dosage , Inflammation Mediators/administration & dosage , Inflammation Mediators/immunology , Vaccines, DNA/immunology , Adoptive Transfer , Animals , Asthma/immunology , Asthma/pathology , Asthma/prevention & control , Bronchial Hyperreactivity/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cations/administration & dosage , Cations/metabolism , Cytokines/physiology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , H-2 Antigens/immunology , Humans , Immunosuppressive Agents/immunology , Liposomes , Methacholine Chloride/administration & dosage , Methacholine Chloride/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
8.
J Immunol ; 183(1): 370-80, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542448

ABSTRACT

Immunodominance limits the TCR diversity of specific antiviral CD8 T cell responses elicited by vaccination or infection. To prime multispecific T cell responses, we constructed DNA vaccines that coexpress chimeric, multidomain Ags (with CD8 T cell-defined epitopes of the hepatitis B virus (HBV) surface (S), core (C), and polymerase (Pol) proteins and/or the OVA Ag as stress protein-capturing fusion proteins. Priming of mono- or multispecific, HLA-A*0201- or K(b)-restricted CD8 T cell responses by these DNA vaccines differed. K(b)/OVA(257-264)- and K(b)/S(190-197)-specific CD8 T cell responses did not allow priming of a K(b)/C(93-100)-specific CD8 T cell response in mice immunized with multidomain vaccines. Tolerance to the S- Ag in transgenic Alb/HBs mice (that express large amounts of transgene-encoded S- Ag in the liver) facilitated priming of subdominant, K(b)/C(93-100)-specific CD8 T cell immunity by multidomain Ags. The "weak" (i.e., easily suppressed) K(b)/C(93-100)-specific CD8 T cell response was efficiently elicited by a HBV core Ag-encoding vector in 1.4HBV-S(mut) tg mice (that harbor a replicating HBV genome that produces HBV surface, core, and precore Ag in the liver). K(b)/C(93-100)-specific CD8 T cells accumulated in the liver of vaccinated 1.4HBV-S(mut) transgenic mice where they suppressed HBV replication. Subdominant epitopes in vaccines can hence prime specific CD8 T cell immunity in a tolerogenic milieu that delivers specific antiviral effects to HBV-expressing hepatocytes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/genetics , Hepatitis B Vaccines/genetics , Hepatitis B Vaccines/immunology , Immunodominant Epitopes/genetics , Lymphocyte Activation/immunology , Vaccines, DNA/immunology , Virus Replication/immunology , Animals , Antigens, Polyomavirus Transforming/administration & dosage , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Line , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/metabolism , Female , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , H-2 Antigens/metabolism , HSP70 Heat-Shock Proteins/administration & dosage , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , Hepatitis B Core Antigens/genetics , Hepatitis B Core Antigens/immunology , Hepatitis B Core Antigens/metabolism , Hepatitis B Vaccines/administration & dosage , Hepatitis B e Antigens/genetics , Hepatitis B e Antigens/immunology , Hepatitis B e Antigens/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/metabolism , Liver Diseases/immunology , Liver Diseases/prevention & control , Liver Diseases/virology , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , Protein Structure, Tertiary/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Virus Replication/genetics
9.
J Immunol ; 182(1): 309-18, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109162

ABSTRACT

Alloimmunity to mismatched donor HLA-Ags and autoimmunity to self-Ags have been hypothesized to play an important role in immunopathogenesis of chronic rejection of transplanted organs. However, it is not known what role, if any, alloimmune response plays in inducing autoimmunity. To test whether Ab-developed posttransplantation to mismatched donor MHC induces autoimmunity and chronic rejection, we developed a murine model wherein anti-MHC class I Abs or control (C1.18.4/anti-keratin) were administered intrabronchially into native lungs. Animals receiving anti-MHC class I, but not control Abs, developed marked cellular infiltration around vessels and bronchiole of lung by day 15, followed by epithelial hyperplasia, fibrosis, and occlusion of the distal airways similar to chronic rejection following human lung transplantation. Lungs of mice receiving anti-MHC class I showed increased expression of chemokines, their receptors, and growth factors, and induced IL-17 as well as de novo Abs to self-Ags, K-alpha1 tubulin, and collagen V. IL-17 neutralization by anti-IL-17 resulted in reduction of autoantibody and lesions induced by anti-MHC class I Abs. Thus, our results indicate that Abs to donor MHC can induce autoimmunity, mediated by IL-17, which plays a pivotal role in chronic rejection postlung transplantation. Therefore, approaches to prevent autoimmunity should be considered for the treatment of chronic rejection postlung transplantation.


Subject(s)
Autoantibodies/administration & dosage , Autoantibodies/physiology , Autoimmune Diseases/immunology , Bronchiolitis Obliterans/immunology , Graft Rejection/immunology , H-2 Antigens/immunology , Animals , Autoantibodies/metabolism , Autoimmune Diseases/pathology , Binding Sites, Antibody , Bronchiolitis Obliterans/pathology , Cell Movement/immunology , Chronic Disease , Fibrosis , Graft Rejection/pathology , H-2 Antigens/administration & dosage , H-2 Antigens/metabolism , Hyperplasia , Immunity, Cellular , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Interleukin-17/physiology , Intubation, Intratracheal , Lung Transplantation/adverse effects , Lung Transplantation/immunology , Lung Transplantation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Syndrome
10.
Immunology ; 115(4): 484-94, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16011517

ABSTRACT

We and others have previously shown that TAP1-/- mice (H-2b) reject grafts from donors without major histocompatibility complex (MHC) disparity that express wild-type levels of H-2b class I molecules (C57BL/6, TAP1+/+ mice). In this same model, we also showed that subcutaneous priming of TAP1-/- mice with synthetic peptides derived from the H-2Kb molecule accelerated graft rejection and that in vivo depletion of CD4+ T cells induced a significant prolongation of graft survival, suggesting an important role for CD4 T cells. We hypothesize that, in this model, rejection is triggered by the recognition of class I molecules or derived peptides, in an inflammatory microenvironment, by a functionally altered autoreactive T-cell repertoire that escapes the control of peripheral regulatory mechanisms. In the present study, we analysed the cellular autoreactivity induced by synthetic peptides derived from the H-2Kb sequence in naive and TAP1-/- mice transplanted with C57BL/6 grafts, and investigated whether intravenous modulation of autoreactivity to these peptides induced transplantation tolerance. We showed that TAP1-/- mice have peripheral autoreactive T cells that recognize H-2Kb peptides. A significant amplification of proliferation against these peptides was detected in TAP1-/- mice that rejected grafts, indicating that the inflammatory context of transplantation induced peripheral expansion of these autoreactive T cells. Furthermore, intravenous injection of H-2Kb-derived peptides significantly prolonged graft survival in some animals. In these mice (> 100 days graft survival), we observed intragraft inhibition of interferon-gamma and interleukin-10 expression, suggesting that these cytokines have an active role during the rejection. In conclusion, our present data indicate that inflammatory autoreactive T cells directed against H-2Kb peptides can be inhibited in the periphery to prolong graft survival in TAP1-/- mice.


Subject(s)
ATP-Binding Cassette Transporters/immunology , Autoimmunity/immunology , Graft Rejection/immunology , H-2 Antigens/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 2 , Animals , Cell Division/immunology , Cells, Cultured , Graft Survival/immunology , H-2 Antigens/immunology , Histocompatibility Antigens Class I/immunology , Injections, Intravenous , Interleukin-10/immunology , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred C57BL , Models, Animal , Peptides/immunology , Spleen/cytology , T-Lymphocytes/immunology
11.
J Immunol ; 172(3): 1444-8, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734720

ABSTRACT

Donor hemopoietic cell engraftment is considered to be an indicator of allograft tolerance. We depleted chimerism with cells specifically presensitized to the bone marrow donor to investigate its role in mixed chimera-induced tolerance. Three experimental models were used: model A, B10.A cells presensitized to B6 (a anti-b cells) were injected into (B6 x D2)F(1) --> B10.A mixed chimeras grafted with DBA/2 skin; model B, anti-B6 presensitized cells prepared in DBA/2 --> B10.A mixed chimeras, thus unresponsive to DBA/2 (a anti-b/tol-d cells), were injected into (B6 x D2)F(1) --> B10.A mixed chimeras grafted with DBA/2 skin; and model C, (BALB/c x B6)F(1) cells presensitized to CBA (d/b anti-k cells) were injected into (B6 x CBA)F(1) --> BALB/c mixed chimeras grafted with B6 skin. Skin was grafted on day 30. Injection of each cell type before skin grafting abolished hemopoietic cell engraftment and prevented allograft acceptance. Injection of presensitized cells after skin grafting resulted in different outcomes depending on the models. In model A, injection of a anti-b cells completely depleted chimerism and caused allograft rejection. In model B, injection of a anti-b/tol-d cells markedly reduced, but did not deplete, peripheral chimerism and maintained skin allograft survival. In model C, d/b anti-k cells reduced chimerism to the background levels but failed to cause graft rejection, probably due to persistence of injected cells which share MHC with skin grafts. Together, the results show that presence of chimeric donor cells is essential in both the induction and maintenance phases of tolerance induced by mixed chimerism.


Subject(s)
Bone Marrow Transplantation/immunology , Radiation Chimera/immunology , Skin Transplantation/immunology , Transplantation Tolerance/genetics , Animals , Bone Marrow Transplantation/methods , Cytotoxicity Tests, Immunologic , Cytotoxicity, Immunologic/genetics , Female , Graft Rejection/genetics , Graft Rejection/immunology , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Radiation Chimera/genetics , Skin Transplantation/methods , Species Specificity , Spleen/cytology , Spleen/immunology , Spleen/transplantation , Transplantation Tolerance/immunology
12.
J Immunol ; 172(1): 699-708, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14688383

ABSTRACT

Thymic selection is controlled by the interaction between TCR and MHC/peptide. Strength and quality of the signal determine whether thymocytes are selected or deleted. The factors that contribute to this signal remain poorly defined. Here we show that fetal thymic organ cultures (FTOCs) derived from OT-I transgenic mice (the OT-I TCR is restricted by K(b)-SIINFEKL) on a K(b)D(b-/-) background support positive selection, but only when provided with soluble H-2K(b)-SIINFEKL complexes. Selection of CD8 T cells is independent of the valency of the ligand or its capability to coengage CD8 molecules. Both CD8alphaalpha and CD8alphabeta T cells are selected by H-2K(b)-SIINFEKL, but only CD8alphabeta cells are capable of releasing IFN-gamma in response to the same ligand. The alpha(4)beta(7) integrin is up-regulated on postselection thymocytes from FTOCs. After adoptive transfer, FTOC-derived OT-I CD8 T cells divide in response to the agonist peptide SIINFEKL. These results establish that CD8 T cells responsive to their nominal peptide-Ag can be generated in FTOC supplemented with soluble MHC class I molecules equipped with the same peptide.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , H-2 Antigens/immunology , Lymphocyte Activation/immunology , Peptide Fragments/immunology , Thymus Gland/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Division/genetics , Cell Division/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Egg Proteins/immunology , Fetus , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , Immunophenotyping , Integrins/biosynthesis , Interferon-gamma/biosynthesis , Interphase/genetics , Interphase/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peyer's Patches/cytology , Peyer's Patches/immunology , Thymus Gland/cytology , Thymus Gland/transplantation
13.
J Immunol ; 171(10): 5554-61, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14607963

ABSTRACT

The phenomenon of tolerance to noninherited maternal Ags (NIMA) is poorly understood. To analyze the NIMA effect C57BL/6 (H-2(b/b)) males were mated with B6D2F(1) (H-2(b/d)) females, whereby 50% of the offspring are H-2(b/b) mice that have been exposed to maternal H-2(d) alloantigens. Controls were H-2(b/b) offspring of C57BL/6 mothers, either inbred C57BL/6 mice or F(1) backcross mice from breedings with H-2(b/d) fathers. We found that 57% of the H-2(b/b) offspring of semiallogeneic (H-2(b/d)) mothers accepted fully allogeneic DBA/2 (H-2(d/d)) heart grafts for >180 days, while similar transplants were all rejected by day 11 in controls (p < 0.0004). Foster nursing studies showed that both oral and in utero exposure to NIMA are required for this tolerogenic effect. An effect of NIMA was also found to extend the survival of skin grafts from a semiallogeneic donor (p < 0.02). Pretransplant analysis of splenocytes showed a 40-90% reduction of IL-2-, IL-5-, and IFN-gamma-producing T cells responding to H-2(d)-expressing APC in NIMA(d)-exposed vs control mice. Injection of pregnant BALB/c-dm2 (H-2L(d)-negative) female mice i.v. with H-2L(d)(61-80) peptide profoundly suppressed the offspring's indirect pathway alloreactive CD4(+) T cell response to H-2L(d). These results suggest that the natural exposure of the fetus and newborn to maternal cells and/or soluble MHC Ags suppresses NIMA-allospecific T cells of the offspring, predisposing to organ transplant tolerance in adult mice.


Subject(s)
H-2 Antigens/immunology , Immune Tolerance , Maternal-Fetal Exchange/immunology , Animals , Animals, Newborn/immunology , Coronary Vessels/transplantation , Crosses, Genetic , Female , Fetus/immunology , Graft Enhancement, Immunologic/methods , Graft Survival/genetics , Graft Survival/immunology , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , Heart Transplantation/immunology , Heart Transplantation/pathology , Histocompatibility Antigen H-2D , Immune Tolerance/genetics , Immunoglobulin G/biosynthesis , Injections, Intravenous , Male , Maternal-Fetal Exchange/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Pregnancy , Skin Transplantation/immunology , T-Lymphocyte Subsets/immunology
14.
J Immunol ; 171(1): 134-41, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12816991

ABSTRACT

Pretransplant infusion of lymphocytes that express a single allogeneic MHC class I Ag has been shown to induce tolerance to skin and heart allografts that express the same alloantigens. In this study, we demonstrate that reconstitution of immunoincompetent mice with spleen cells from MHC class I L(d)-mismatched donors does not cause graft-vs-host disease (GVHD). Recipient mice become tolerant to skin allografts of lymphocyte donor origin while retaining immunity to third-party alloantigens. The mechanism involves donor-derived CD3(+)CD4(-)CD8(-) double-negative T regulatory (DN Treg) cells, which greatly increase and form the majority of T lymphocytes in the spleen of recipient mice. DN Treg cells isolated from tolerant recipient mice can suppress the proliferation of syngeneic antihost CD8(+) T cells in vitro. Furthermore, we demonstrate that DN Treg cells can be generated in vitro by stimulating them with MHC class I L(d)-mismatched lymphocytes. These in vitro generated L(d)-specific DN Treg cells are able to down-regulate the activity of antihost CD8(+) T cells in vitro by directly killing activated CD8(+) T cells. Moreover, infusing in vitro generated L(d)-mismatched DN Treg cells prevented the development of GVHD caused by allogeneic CD8(+) T cells. Together these data demonstrate that infusion of single MHC class I locus-mismatched lymphocytes may induce donor-specific transplantation tolerance through activation of DN Treg cells, which can suppress antihost CD8(+) T cells and prevent the development of GVHD. This finding indicates that using single class I locus-mismatched grafts may be a viable alternative to using fully matched grafts in bone marrow transplantation.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD3 Complex/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Division/genetics , Cell Division/immunology , Clone Cells , Coculture Techniques , Cytotoxicity, Immunologic/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Histocompatibility Testing , Lymphocyte Transfusion , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Skin Transplantation/adverse effects , Skin Transplantation/immunology , Spleen/cytology , Spleen/immunology , Spleen/transplantation , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Transplantation Tolerance/genetics , Transplantation, Isogeneic/immunology , Transplantation, Isogeneic/pathology
15.
J Immunol ; 170(1): 228-35, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12496404

ABSTRACT

Tumor membrane Ag immobilized on cell size microspheres (large multivalent immunogen (LMI)) was previously shown to augment tumor-specific CTL activity and reduce tumor growth, and a clinical trial examining this approach is in progress. In the current study, LMI treatment has been examined using adoptive transfer of TCR-transgenic CD8 T cells to visualize Ag-specific cells during the response. OT-I T cells specific for H-2K(b)/OVA(257-264) were transferred into mice that were then challenged with LMI made by immobilizing H-2K(b)/OVA(257-264) on microspheres (K(b)/OVA(257-264)-LMI) alone, or along with i.p. challenge with OVA-expressing E.G7 tumor. K(b)/OVA(257-264)-LMI caused significant reduction of tumor growth when administered to E.G7-bearing mice. When administered alone, the K(b)/OVA(257-264)-LMI caused only weak clonal expansion of OT-I cells in the spleen and lymph nodes, although most of the OT-I cells up-regulated expression of CD44 and VLA-4. In contrast, K(b)/OVA(257-264)-LMI administration to E.G7-bearing mice stimulated no detectable expansion of OT-I cells in the spleen and lymph nodes but caused a rapid increase in the number of OT-I cells in the peritoneal cavity, the site of the growing tumor. These results demonstrate the potential for using class I/tumor peptide complexes for immunotherapy. In addition, they suggest a model for the mechanism of CTL augmentation in which recognition of the LMI Ag results in altered trafficking of the tumor-specific CD8 T cells so that they reach the site of a growing tumor more rapidly and in greater numbers, where they may further expand and acquire effector function.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/physiology , Antigens, Neoplasm/immunology , Cytotoxicity, Immunologic , Egg Proteins/immunology , H-2 Antigens/physiology , Lymphocyte Activation/immunology , Ovalbumin/immunology , T-Lymphocytes, Cytotoxic/transplantation , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Egg Proteins/administration & dosage , Growth Inhibitors/administration & dosage , Growth Inhibitors/pharmacology , H-2 Antigens/administration & dosage , H-2 Antigens/immunology , Immunodominant Epitopes/physiology , Injections, Intraperitoneal , Injections, Intravenous , Macromolecular Substances , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microspheres , Neoplasm Transplantation , Ovalbumin/administration & dosage , Peptide Fragments , T-Lymphocytes, Cytotoxic/immunology , Thymoma/immunology , Thymoma/prevention & control , Tumor Cells, Cultured
16.
Hum Immunol ; 63(10): 888-92, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12368041

ABSTRACT

We have previously reported that intratracheal delivery of donor splenocytes in mice induces hyporesponsiveness to fully allogeneic cardiac grafts and generates regulatory cells. Here, we examined whether an allopeptide would produce the same results. A 15-mer (54-68) peptide corresponding to a hypervariable region of the K(b) molecule was given intratracheally or intravenously to CBA (H2(k)) mice 7 days before transplantation of a C57BL/10 (H2(b)) or BALB/c (H2(d)) heart and was also used in adoptive transfer experiments. Cardiac grafts in recipients given K(b) peptide intratracheally experienced a median survival time (MST) of 56 days, whereas those in recipients given the peptide intravenously were rejected acutely (MST=7.5 days). Adoptive transfer of splenocytes from mice pretreated intratracheally with K(b) peptide to naïve secondary recipients prolonged survival of cardiac grafts (MST = 35 days). Intratracheal delivery of a single major histocompatibility complex class I peptide induced hyporesponsiveness to allogeneic cardiac grafts and generated regulatory cells.


Subject(s)
Graft Survival , H-2 Antigens/administration & dosage , Heart Transplantation/immunology , Adoptive Transfer , Animals , Immune Tolerance , Interferon-gamma/biosynthesis , Mice , Mice, Inbred Strains , Trachea , Transplantation, Homologous
17.
J Immunol ; 169(6): 2937-46, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12218107

ABSTRACT

Substantial effort has been invested into optimization of vector structure, DNA formulation, or delivery methods to increase the effectiveness of DNA vaccines. In contrast, it has been only insufficiently explored how the higher order structure of an antigenic protein influences immunogenicity of embedded epitopes in vivo. Potent CD8+ T cell responses specific for a single immunogenic epitope are induced upon electrovaccination with plasmid DNA encoding the full-length heavy chain of the human HLA-Cw3 molecule. Contrary to expectations, a minimal construct, which provoked a substantial release of IFN-gamma from specific CTLs in vitro, did not induce a significant response in vivo. Systematically altered variants of the Cw3 molecule were thus tested both in vivo and in vitro to determine which structural parts are responsible for this discrepancy. In complementation experiments the participation of trans-acting helper epitopes was ruled out. Successive C-terminal truncations, human/mouse domain swap variants, and subdomain modifications defined the alpha3 region of the HLA heavy chain and membrane anchoring as critical elements. Based on these data, refined minimal constructs were engineered that triggered very high in vivo responses. The most advanced variant consisted only of an adenoviral leader, antigenic epitope, alpha3 domain, and 16 aa of the transmembrane domain. When a tumor Ag epitope was incorporated into one of these high performer minimal constructs, protection against melanoma metastases was attained upon vaccination. Thus, structural elements of the Ag can dominantly influence immunogenicity in vivo. These elements can also markedly improve the immunogenicity of unrelated Ags and may form the basis of a new generation of DNA vaccines.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , H-2 Antigens/chemistry , H-2 Antigens/immunology , HLA-C Antigens/chemistry , HLA-C Antigens/immunology , Animals , Antigens, Neoplasm/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemical synthesis , Cancer Vaccines/immunology , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/immunology , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/genetics , Female , Genetic Complementation Test , H-2 Antigens/administration & dosage , HLA-C Antigens/administration & dosage , HLA-C Antigens/genetics , Lymphocyte Activation/genetics , Melanoma, Experimental/chemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Plasmids/administration & dosage , Plasmids/chemical synthesis , Plasmids/immunology , Protein Engineering/methods , Protein Structure, Tertiary/genetics , Sequence Deletion , Structure-Activity Relationship , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
18.
J Immunol ; 167(7): 3708-14, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564786

ABSTRACT

Soluble MHC/peptide tetramers that can directly bind the TCR allow the direct visualization and quantitation of Ag-specific T cells in vitro and in vivo. We used HY-D(b) tetramers to assess the numbers of HY-reactive CD8+ T cells in HYTCR-transgenic mice and in naive, wild-type C57BL/6 (B6) mice. As expected, tetramer staining showed the majority of T cells were male-specific CD8+ T cells in female HY-TCR mice. Staining of B6 mice showed a small population of male-specific CD8+ T cells in female mice. The effect of administration of soluble MHC class I tetramers on CD8+ T cell activation in vivo was unknown. Injection of HY-D(b) tetramer in vivo effectively primed female mice for a more rapid proliferative response to both HY peptide and male splenocytes. Furthermore, wild-type B6 female mice injected with a single dose of HY-D(b) tetramer rejected B6 male skin grafts more rapidly than female littermates treated with irrelevant tetramer. In contrast, multiple doses of HY-D(b) tetramer did not further decrease graft survival. Rather, female B6 mice injected with multiple doses of HY-D(b) tetramer rejected male skin grafts more slowly than mice primed with a single injection of tetramer or irradiated male spleen cells, suggesting clonal exhaustion or anergy. Our data highlight the ability of soluble MHC tetramers to identify scarce alloreactive T cell populations and the use of such tetramers to directly modulate an Ag-specific T cell response in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , H-2 Antigens/immunology , H-Y Antigen/immunology , Lymphocyte Activation , Animals , Cells, Cultured , Clonal Anergy , Female , Genes, T-Cell Receptor , Graft Rejection/immunology , Graft Survival , H-2 Antigens/administration & dosage , Histocompatibility Antigen H-2D , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Skin Transplantation
19.
J Immunol ; 164(10): 5492-8, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10799917

ABSTRACT

A conformationally biased decapeptide agonist of human C5a anaphylatoxin (YSFKPMPLaR) was used as a molecular adjuvant in stimulating an Ag-specific CTL response against murine P815S target cells expressing an Ld-restricted CTL epitope of the hepatitis B surface Ag (HBsAg). Groups of BALB/c mice (H-2d) were immunized with aqueous solutions of the HBsAg CTL epitopes (IPQSLDSWWTSL and IPQSLDSWWTSLRR); the C5a agonist (YSFKPMPLaR); the C5a agonist and HBsAg CTL epitopes admixed (IPQSLDSWWTSL and IPQSLDSWWTSLRR + YSFKPMPLaR); the C5a-active, HBsAg CTL epitope-C5a agonist constructs (IPQSLDSWWTSLYSFKPMPLaR, IPQSLDSWWTSLRRYSFKPMPLaR, and IPQSLDSWWTSLRVRRYSFPMPLaR); a C5a-inactive, reverse-moiety construct (YSFKPMPLaRRRIPQSLDSWWTSL); and a C5a-attenuated, carboxyl-terminal-blocked construct (IPQSLDSWWTSLRRYSFKPMPLaRG). Ag-specific CD8+ CTL responses were observed after the secondary boost in the absence of any added adjuvant only in mice that were immunized with C5a-active contructs, IPQSLDSWWTSLRRYSFKPMPLaR and IPQSLDSWWTSLRVRRYSFKPMPLaR. These two C5a-active immunogens contained potential subtilisin-sensitive linker sequences between the HBsAg CTL epitope and the C5a agonist; i.e., a double-Arg (RR) and a furin protease sensitive sequence (RVRR). The introduction of these potentially cleavable sequences may be a method of increasing the likelihood of liberating the CTL epitope from the C5a agonist by intracellular proteases, thereby facilitating entry of the epitope into Ag-processing pathways via an exogenous route.


Subject(s)
Adjuvants, Immunologic/agonists , Adjuvants, Immunologic/chemistry , Complement C5a/agonists , Complement C5a/chemistry , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/immunology , T-Lymphocytes, Cytotoxic/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Amino Acid Sequence , Animals , Arginine/administration & dosage , Arginine/chemistry , Arginine/immunology , Cells, Cultured , Complement C5a/administration & dosage , Complement C5a/immunology , Endopeptidases/chemistry , Endopeptidases/immunology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/chemistry , Female , H-2 Antigens/administration & dosage , H-2 Antigens/chemistry , H-2 Antigens/immunology , Hepatitis B Antibodies/biosynthesis , Hepatitis B Surface Antigens/administration & dosage , Hepatitis B Surface Antigens/immunology , Histocompatibility Antigen H-2D , Humans , Injections, Subcutaneous , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Protein Conformation
20.
Eur J Immunol ; 28(8): 2395-406, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9710217

ABSTRACT

Peripheral tolerance is considered to be a safeguard against autoimmunity. Using a TCR-transgenic mouse system displaying peripheral tolerance against a liver-specific MHC class I Kb antigen, we investigated whether the breaking of tolerance would result in autoimmunity. Reversal of tolerance was achieved by simultaneous challenge with cells expressing the Kb autoantigen and IL-2. Tolerance could not be broken with IL-2 alone or when Kb- and IL-2-expressing cells were applied to different sites of the mice. However, despite the presence of activated autoreactive T cells that were able to reject Kb-positive grafts no autoaggression against the Kb-positive liver was observed. These results indicate that breaking of tolerance per se is not sufficient to cause liver-specific autoimmunity. However, when in addition to breaking tolerance the mice were infected with a liver-specific pathogen, autoaggression occurred. Thus, in this system at least two independent steps seem to be required for organ-specific autoimmunity: reversal of peripheral tolerance resulting in functional activation of autoreactive T cells and conditioning of the liver microenvironment which enables the activated T cells to cause tissue damage.


Subject(s)
Autoimmunity , Self Tolerance , Animals , Autoantigens/administration & dosage , Autoantigens/genetics , Graft Rejection/immunology , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , Hepatitis, Animal/immunology , Hepatitis, Animal/pathology , Interleukin-2/pharmacology , Listeriosis/immunology , Listeriosis/pathology , Liver/immunology , Lymphocyte Activation , Mast-Cell Sarcoma/immunology , Mice , Mice, Inbred CBA , Mice, Inbred DBA , Mice, Transgenic , Neoplasm Transplantation , Organ Specificity , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...