Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Immunol Lett ; 192: 61-71, 2017 12.
Article in English | MEDLINE | ID: mdl-29111199

ABSTRACT

Peripheral T cell tolerance is challenging to induce in partially lymphopenic hosts and this is relevant for clinical situations involving transplant tolerance. While the shortage of regulatory cells is thought to be one reason for this, T cell-intrinsic tolerance processes such as anergy are also poorly triggered in such hosts. In order to understand the latter, we used a T cell deficient mouse model system where adoptively transferred autoreactive T cells are significantly tolerized in a cell intrinsic fashion, without differentiation to regulatory T cells. Intriguingly these T cells often retain sufficient effector functions to trigger autoimmune pathology. Here we find that the high population density of the autoreactive T cells that accumulated in such a host limits the progression of the cell-intrinsic tolerance process in T cells. Accordingly, reducing the cell density during a second transfer allowed T cells to further tune down their responsiveness to antigenic stimulation. The retuning of T cells was reflected by a loss of the T cell's abilities to proliferate, produces cytokines or help B cells. We further suggest, based on altering the levels of chronic antigen using miniosmotic pumps, that the effects of cell-density on T cell re-tuning may reflect the effective changes in the antigen dose perceived by individual T cells. This could proportionally elicit more negative feedback downstream of the TCR. Consistent with this, the retuned T cells showed signaling defects both proximal and distal to the TCR. Therefore, similar to the immunogenic activation of T cells, cell-intrinsic T cell tolerance may also involve a quantitative and progressive process of tuning down its antigen-responsiveness. The progress of such tuning seems to be stabilized at multiple intermediate stages by factors such as cell density, rather than just absolute antigen levels.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphopenia/immunology , Peripheral Tolerance , Adoptive Transfer , Animals , Autoantigens/immunology , Autoimmunity , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , DNA-Binding Proteins/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Nat Commun ; 6: 8698, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26507712

ABSTRACT

Induction of T-cell clonal anergy involves serial activation of transcription factors, including NFAT and Egr2/3. However, downstream effector mechanisms of these transcription factors are not fully understood yet. Here we identify Ndrg1 as an anergy factor induced by Egr2. Ndrg1 is upregulated by anergic signalling and maintained at high levels in resting anergic T cells. Overexpression of Ndrg1 mimics the anergic state and knockout of the gene prevents anergy induction. Interestingly, Ndrg1 is phosphorylated and degraded by CD28 signalling in a proteasome-dependent manner, explaining the costimulation dependence of anergy prevention. Similarly, IL-2 treatment of anergic T cells, under conditions that lead to the reversal of anergy, also induces Ndrg1 phosphorylation and degradation. Finally, older Ndrg1-deficient mice show T-cell hyperresponsiveness and Ndrg1-deficient T cells aggravate inducible autoimmune inflammation. Thus, Ndrg1 contributes to the maintenance of clonal anergy and inhibition of T-cell-mediated inflammation.


Subject(s)
CD28 Antigens/immunology , Cell Cycle Proteins/genetics , Clonal Anergy , Down-Regulation , Interleukin-2/immunology , Intracellular Signaling Peptides and Proteins/genetics , T-Lymphocytes/immunology , Animals , CD28 Antigens/genetics , Cell Cycle Proteins/immunology , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/immunology , Interleukin-2/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Immunity ; 38(6): 1198-210, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23809163

ABSTRACT

Differences in gut commensal flora can dramatically influence autoimmune responses, but the mechanisms behind this are still unclear. We report, in a Th1-cell-driven murine model of autoimmune arthritis, that specific gut commensals, such as segmented filamentous bacteria, have the ability to modulate the activation threshold of self-reactive T cells. In the local microenvironment of gut-associated lymphoid tissues, inflammatory cytokines elicited by the commensal flora dynamically enhanced the antigen responsiveness of T cells that were otherwise tuned down to a systemic self-antigen. Together with subtle differences in early lineage differentiation, this ultimately led to an enhanced recruitment of pathogenic Th1 cells and the development of a more severe form of autoimmune arthritis. These findings define a key role for the gut commensal flora in sustaining ongoing autoimmune responses through the local fine tuning of T-cell-receptor-proximal activation events in autoreactive T cells.


Subject(s)
Arthritis, Experimental/immunology , Autoimmune Diseases/immunology , Clostridium/immunology , Intestines/immunology , Th1 Cells/immunology , Animals , Arthritis, Experimental/microbiology , Autoantigens/immunology , Autoimmune Diseases/microbiology , CD3 Complex/genetics , Cells, Cultured , Clonal Selection, Antigen-Mediated , Disease Models, Animal , Humans , Intestines/microbiology , Lymphocyte Activation , Metagenome , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Th1 Cells/microbiology
4.
Immunity ; 37(4): 735-46, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23021952

ABSTRACT

After an immune response, the expanded population of antigen-specific CD4(+) T cells contract to steady state levels. We have found that the contraction is neither cell-autonomous nor mediated by competition for generic trophic factors, but regulated by relatively rare subsets of neighboring CD4(+) T cells not necessarily of a conventional regulatory T cell lineage. These regulators, referred to as deletors, specifically limit the frequency of particular antigen-specific T cells even though they are not reactive to the same agonist as their targets. Instead, an isolated deletor could outcompete the target for recognition of a shared, nonstimulatory endogenous peptide-MHC ligand. This mechanism was sufficient to prevent even agonist-driven autoimmune disease in a lymphopenic environment. Such a targeted regulation of homeostasis within narrow colonies of T cells with related TCR specificities for subthreshold ligands might help to prevent the loss of unrelated TCRs during multiple responses, preserving the valuable diversity of the repertoire.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , T-Cell Antigen Receptor Specificity , Animals , Autoimmunity , Cell Survival , Cells, Cultured , Ligands , Lymphopenia/immunology , Mice , Receptors, Antigen, T-Cell/immunology
5.
Eur J Immunol ; 42(11): 2875-80, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22806568

ABSTRACT

The behavior of self-reactive T cells in the peripheral immune system has often been studied by following the fate of adoptively transferred antigen-specific T cells in antigen expressing mice. In most cases, after a period of expansion, such cells undergo a slow clonal deletion, accompanied by the onset of anergy and/or suppression in the remaining cells. Here, we demonstrate that at initial frequencies approaching those found in normal repertoires, it is possible to completely avoid deletion and still maintain peripheral tolerance. At starting numbers of <1000 T cells, stimulation by chronic self-antigens resulted in a period of robust clonal expansion, followed by a steady plateau phase extending beyond 4 months. Despite their stable persistence, the self-reactive T cells did not convert to a Foxp3⁺ fate. However, they displayed a considerable block in their ability to make IL-2, consistent with the onset of anergy - in a precursor frequency or deletion independent fashion.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Clonal Anergy/immunology , Adoptive Transfer , Animals , Flow Cytometry , Immune Tolerance/immunology , Interleukin-2/immunology , Male , Mice , Mice, Knockout , Mice, Transgenic , Specific Pathogen-Free Organisms
6.
Transplantation ; 94(3): 234-40, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22797707

ABSTRACT

BACKGROUND: The hyperacute rejection mediated by preexisting antibodies is a major impediment to the success of transplants across allogeneic and xenogeneic barriers. We report a new mouse model that allows us to not only monitor the sensitization of B cells mediating the hyperacute response but also validate therapeutic strategies for tolerizing them. MODEL: The new model system uses 5C.C7,RAG2 T-cell receptor transgenic T cells and B10.S(9R),CD3[Latin Small Letter Open E] hosts for adoptive transfer experiments. RESULTS AND CONCLUSIONS: In the allogeneic hosts, transgenic T cells expanded briefly before being chronically deleted. Once the deletion was initiated, a second graft of donor cells was used to assess a hyperacute response. The rapid rejection of the second cohort correlated with the appearance of donor-specific antibodies in the serum. Interestingly, chronically stimulated T cells were relatively resistant to hyperacute rejection, suggesting an explanation for the slower rejection kinetics of the first cohort even as the second cohort of identical donor cells was being hyperacutely rejected. Finally, we could tolerize the potential for a hyperacute response, by pretreating recipients with a single infusion of naive donor B cells before the first T-cell transfer. This treatment not only abrogated the development of a hyperacute response but also allowed the primary graft to survive in vivo for extended periods.


Subject(s)
Disease Models, Animal , Graft Rejection/immunology , Graft vs Host Reaction , Alleles , Animals , B-Lymphocytes/cytology , Cell Separation , Cohort Studies , Crosses, Genetic , Flow Cytometry , Graft Survival , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/cytology
7.
Cold Spring Harb Perspect Biol ; 4(4): a006908, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22395097

ABSTRACT

A fundamental property of the immune system is its ability to mediate self-defense with a minimal amount of collateral damage to the host. The system uses several different mechanisms to achieve this goal, which is collectively referred to as the "process of immunological tolerance." This article provides an introductory historical overview to these various mechanisms, which are discussed in greater detail throughout this collection, and then briefly describes what happens when this process fails, a state referred to as "autoimmunity."


Subject(s)
Immune Tolerance , Humans
8.
J Immunol ; 187(6): 3292-9, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21832163

ABSTRACT

Most gene promoters have multiple binding sequences for many transcription factors, but the contribution of each of these factors to chromatin remodeling is still unclear. Although we previously found a dynamic change in the arrangement of nucleosome arrays at the Il2 promoter during T cell activation, its timing preceded that of a decrease in nucleosome occupancy at the promoter. In this article, we show that the initial nucleosome rearrangement was temporally correlated with the binding of NFAT1 and AP-1 (Fos/Jun), whereas the second step occurred in parallel with the recruitment of other transcription factors and RNA polymerase II. Pharmacologic inhibitors for activation of NFAT1 or induction of Fos blocked the initial phase in the sequential changes. This step was not affected, however, by inhibition of c-Jun phosphorylation, which instead blocked the binding of the late transcription factors, the recruitment of CREB-binding protein, and the acetylation of histone H3 at lysine 27. Thus, the sequential recruitment of transcription factors appears to facilitate two separate steps in chromatin remodeling at the Il2 locus.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Interleukin-2/genetics , Lymphocyte Activation/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Animals , Blotting, Western , CREB-Binding Protein/genetics , CREB-Binding Protein/immunology , CREB-Binding Protein/metabolism , Chromatin/genetics , Chromatin/immunology , Chromatin/metabolism , Chromatin Assembly and Disassembly/immunology , Chromatin Immunoprecipitation , Female , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocytes/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Transcription Factor AP-1/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism
9.
J Immunol ; 187(2): 805-16, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21685322

ABSTRACT

Adaptive tolerance is a hyporesponsive state in which lymphocyte Ag receptor signaling becomes desensitized after prolonged in vivo encounter with Ag. The molecular mechanisms underlying this hyporesponsive state in T cells are not fully understood, although a major signaling block has been shown to be present at the level of ZAP70 phosphorylation of linker for activation of T cells (LAT). In this study, we investigated the ability of adaptively tolerant mouse T cells to form conjugates with Ag-bearing APCs and to translocate signaling molecules into the interface between the T cells and APCs. Compared with naive or preactivated T cells, adaptively tolerant T cells showed no dramatic impairment in their formation of conjugates with APCs. In contrast, there was a large impairment in immunological synapse formation. Adaptively tolerant T cells were defective in their translocation of signaling molecules, such as ZAP70, LAT, and phospholipase C γ1, into the T cell-APC contact sites. Although Ag-induced activation of VAV1 was normal, VAV's recruitment into the synapse was also impaired. Interestingly, expressions of both IL-2-inducible T cell kinase and growth factor receptor-bound protein 2-related adaptor downstream of SHC were decreased by 60-80% in adaptively tolerant T cells. These decreases, in addition to the impairment in LAT phosphorylation by ZAP70, appear to be the major impediments to the phosphorylation of SLP76 (SRC homology 2 domain-containing leukocyte protein of 76 kDa) and the recruitment of VAV1, which are important for stable immunological synapse formation.


Subject(s)
Adaptive Immunity , Clonal Anergy , Down-Regulation/immunology , Immunological Synapses , T-Lymphocyte Subsets/immunology , Adaptive Immunity/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Line , Clonal Anergy/genetics , Down-Regulation/genetics , GRB2 Adaptor Protein/antagonists & inhibitors , Immunological Synapses/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphorylation/immunology , Protein Transport/immunology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins c-vav , T-Lymphocyte Subsets/metabolism
10.
J Exp Med ; 207(11): 2521-32, 2010 Oct 25.
Article in English | MEDLINE | ID: mdl-20937703

ABSTRACT

Niche availability provided by stromal cells is critical to thymus function. Thymi with diminished function contain fewer stromal cells, whereas thymi with robust function contain proliferating stromal cell populations. Here, we show that the thymus, brain, and testes-associated gene (Tbata; also known as SPATIAL) regulates thymic epithelial cell (TEC) proliferation and thymus size. Tbata is expressed in thymic stromal cells and interacts with the enzyme Uba3, thereby inhibiting the Nedd8 pathway and cell proliferation. Thymi from aged Tbata-deficient mice are larger and contain more dividing TECs than wild-type littermate controls. In addition, thymic reconstitution after bone marrow transplantation occurred more rapidly in Rag2(-/-)Tbata(-/-) mice than in Rag2(-/-)Tbata(+/+) littermate controls. These findings suggest that Tbata modulates thymus function by regulating stromal cell proliferation via the Nedd8 pathway.


Subject(s)
Nuclear Proteins/metabolism , Thymus Gland/immunology , Ubiquitins/metabolism , Aging/genetics , Aging/immunology , Aging/metabolism , Animals , Bone Marrow Transplantation/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Humans , Mice , Mice, Knockout , NEDD8 Protein , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Stromal Cells/cytology , Stromal Cells/immunology , Stromal Cells/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transplantation, Homologous , Ubiquitins/genetics , Ubiquitins/immunology
11.
Curr Opin Immunol ; 22(5): 552-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20869863

ABSTRACT

Anergy is a state of long-term hyporesponsiveness in T cells that is characterized by an active repression of TCR signaling and IL-2 expression [1]. Several forms of anergy have been described and the past few years have brought to light an increasing number of 'anergic factors' involved in the induction and the active maintenance of the state in lymphocytes. The role of mTOR and other related metabolic sensors and regulators has recently emerged as of particular importance in broadening our view of anergy-inducing signals. We will discuss the role of these molecules in regulating the choice between anergy and activation, a decision faced by all T cells undergoing TCR stimulation. We will then explore the relationship between the induction of anergy and the induction of regulatory T cells as well as the potential crosstalk responsible for the phenomenon of infectious tolerance.


Subject(s)
Clonal Anergy/immunology , Infections/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Cues , Environmental Exposure , Humans , Lymphocyte Activation/immunology
12.
Nucleic Acids Res ; 38(11): e124, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20371521

ABSTRACT

To explore the higher order structure of transcribable chromatin in vivo, its local configuration was assessed through the accessibility of the chromatin to crosslinking with formaldehyde. The application of crosslinked and mildly sheared chromatin to sedimentation velocity centrifugation followed by size-fractionation of the DNA enabled us to biochemically distinguish between chromatin with heavily versus sparsely crosslinkable structures. The separated fractions showed a good correlation with gene expression profiles. Genes with poor crosslinking around the promoter region were actively transcribed, while transcripts were hardly detected from genes with extensive crosslinking in their promoter regions. For the inducible gene, Il2, the distribution of the promoter shifted in the gradient following T-cell receptor stimulation, consistent with a change in structure at this locus during activation. The kinetics of this switch preceded the chromatin change observed in a DNase I accessibility assay. Thus, this new chromatin fractionation technique has revealed a change in chromatin structure that has not been previously characterized.


Subject(s)
Chromatin/chemistry , Cross-Linking Reagents , Formaldehyde/chemistry , Promoter Regions, Genetic , Ultracentrifugation/methods , Animals , DNA/isolation & purification , Female , Gene Expression Profiling , Interleukin-2/genetics , Lymphocyte Activation , Mice , Mice, Transgenic , Proteins/isolation & purification , T-Lymphocytes/immunology
14.
Nat Immunol ; 10(8): 831-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19597497

ABSTRACT

T cell antigen receptor (TCR) signaling in CD4(+)CD8(+) double-positive thymocytes determines cell survival and lineage commitment, but the genetic and molecular basis of this process is poorly defined. To address this issue, we used ethylnitrosourea mutagenesis to identify a previously unknown T lineage-specific gene, Themis, which is critical for the completion of positive selection. Themis contains a tandem repeat of a unique globular domain (called 'CABIT' here) that includes a cysteine motif that defines a family of five uncharacterized vertebrate proteins with orthologs in most animal species. Themis-deficient thymocytes showed no substantial impairment in early TCR signaling but did show altered expression of genes involved in the cell cycle and survival before and during positive selection. Our data suggest a unique function for Themis in sustaining positive selection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage/physiology , Proteins/physiology , Receptors, Antigen, T-Cell/physiology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Line , Cell Survival/physiology , Ethylnitrosourea/pharmacology , Female , Humans , Intercellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Mutation , Oligonucleotide Array Sequence Analysis , Proteins/genetics , Proteins/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction
15.
J Immunol ; 181(2): 1272-81, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606681

ABSTRACT

We describe a novel biphasic regulation of Il2 transcription in naive CD4(+) T cells. Few ( approximately 5%) CD4(+) T cells transcribe Il2 within 6 h of anti-TCR-beta plus anti-CD28 stimulation (early phase). Most naive CD4(+) T cells do not initiate Il2 transcription until after an additional approximately 12 h of T cell stimulation (late phase). In comparison, essentially all previously activated (Pre-Ac) CD4(+) T cells that transcribe Il2 do so with an early-phase response. Late-phase Il2 expression mostly requires c-Rel, CD28, and TNFR signaling. In contrast, early-phase transcription is only partly c-Rel and CD28 dependent and TNFR independent. There was also increased stable DNA accessibility at the Il2 locus and elevated c-Rel expression in resting Pre-Ac CD4(+) cells. Upon T cell activation, a faster and greater increase in DNA accessibility as well as c-Rel nuclear expression were observed in Pre-Ac CD4(+) cells relative to naive CD4(+) T cells. In addition, both acetylated histone H3 and total H3 decreased at the Il2 locus upon rechallenge of Pre-Ac CD4(+) T cells, whereas increased acetylated histone H3 with no change in total H3 was observed following activation of naive CD4(+) T cells. We propose a model in which nucleosome disassembly facilitates rapid initiation of Il2 transcription in CD4(+) T cells, and suggest that a threshold level of c-Rel must be reached for Il2 promoter activity in both naive and Pre-Ac CD4(+) T cells. This is provided, at least partially, by TNFR signaling during priming, but not during recall.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chromatin/metabolism , Interleukin-2/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism , Acetylation , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Histones/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocyte Activation , Mice , Mice, Mutant Strains , Nucleosomes/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/metabolism , Receptors, Tumor Necrosis Factor, Type I/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/immunology , Up-Regulation
16.
J Immunol ; 179(12): 7999-8008, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18056339

ABSTRACT

The outcome of peripheral T cell activation is thought to be largely determined by the context in which the cognate Ag is initially presented. In this framework, microbial products that can activate APCs via TLRs are considered critical in converting an otherwise tolerogenic context to an immunogenic one. We examine this idea using a model system where naive T cells are stimulated in the periphery by a persistent self Ag. The addition of multiple TLR ligands to this context, acutely or chronically, failed to significantly alter the tolerogenic phenotype in the responding T cells. This contrasts with the ability of such adjuvants to improve T cell responses to soluble peptide immunizations. We reconcile this difference by revealing a hitherto poorly appreciated property of TLR ligands, which extends the duration of soluble Ag presentation in vivo by an additional two to three days. Finally, we could replace the requirement for TLR-mediated APC activation in soluble-Ag-induced T cell expansion and differentiation, by maintaining the Ag depot in vivo using repeated immunizations. These data suggest a novel process by which TLR ligands modulate T cell responses to acute Ags, without disrupting the induction of tolerance to persistent self Ags.


Subject(s)
Antigen Presentation , Autoantigens/immunology , Autoimmune Diseases/immunology , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism , Animals , Immune Tolerance , Ligands , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Mice , Peptides/immunology , Peptides/pharmacology , Toll-Like Receptors/agonists
17.
Semin Immunol ; 19(3): 140-52, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17400472

ABSTRACT

Since the original description of T cell anergy in CD4 clones from mice and humans, a number of different unresponsive states have been described, both in vivo and in vitro, that have been called anergic. While initial attempts were made to understand the similarities between the different models, it has now become clear from biochemical experiments that many of them have different molecular mechanisms underlying their unresponsiveness. In this review we will detail our own work on the in vivo model referred to as adaptive tolerance and then attempt to compare this biochemical state to the multitude of other states that have been described in the literature.


Subject(s)
Clonal Anergy/immunology , Immune Tolerance/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Humans , Mice , T-Lymphocytes/metabolism
18.
Immunity ; 25(6): 851-5, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17174925

ABSTRACT

A recent surge of interest in the behavior of T and B cells in lymphopenic model systems has resurrected a certain cynicism about the validity of using such models to answer important immunological questions. Here we discuss this skepticism in a broader historical context.


Subject(s)
Disease Models, Animal , Lymphopenia , Models, Immunological , Adoptive Transfer , Animals , B-Lymphocytes/radiation effects , Mice , T-Lymphocytes/radiation effects
19.
PLoS Biol ; 4(11): e340, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17048986

ABSTRACT

Overlapping roles have been ascribed for T cell anergy, clonal deletion, and regulation in the maintenance of peripheral immunological tolerance. A measurement of the individual and additive impacts of each of these processes on systemic tolerance is often lacking. In this report we have used adoptive transfer strategies to tease out the unique contribution of T cell intrinsic receptor calibration (adaptation) in the maintenance of tolerance to a systemic self-antigen. Adoptively transferred naïve T cells stably calibrated their responsiveness to a persistent self-antigen in both lymphopenic and T cell-replete hosts. In the former, this state was not accompanied by deletion or suppression, allowing us to examine the unique contribution of adaptation to systemic tolerance. Surprisingly, adapting T cells could chronically help antigen-expressing B cells, leading to polyclonal hypergammaglobulinemia and pathology, in the form of mild arthritis. The helper activity mediated by CD40L and cytokines was evident even if the B cells were introduced after extended adaptation of the T cells. In contrast, in the T cell-replete host, neither arthritis nor autoantibodies were induced. The containment of systemic pathology required host T cell-mediated extrinsic regulatory mechanisms to synergize with the cell intrinsic adaptation process. These extrinsic mechanisms prevented the effector differentiation of the autoreactive T cells and reduced their precursor frequency, in vivo.


Subject(s)
Adaptation, Biological/immunology , Immune Tolerance/physiology , T-Lymphocytes/physiology , Adoptive Transfer , Animals , Arthritis/etiology , Arthritis/immunology , Autoantibodies/physiology , Autoimmune Diseases/immunology , B-Lymphocytes/physiology , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Cell Differentiation/immunology , Cells, Cultured , Clonal Anergy , Cytokines/physiology , Immunosuppression Therapy , Lymphocyte Activation/physiology , Mice , Mice, Transgenic , Protein Binding
20.
Nat Clin Pract Rheumatol ; 2(1): 44-52, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16932651

ABSTRACT

Successful adaptive immunity against a broad range of pathogens depends on the diversity of randomly generated T-lymphocyte and B-lymphocyte receptors. A subset of these receptors will be self-reactive and must be regulated to prevent autoimmunity. The process of immunologic tolerance addresses this problem by either purging autoreactive receptors from the system or tuning down their reactivity sufficiently to prevent disease. Immature lymphocytes generate a novel receptor during development in the thymus or bone marrow. Engagement of self antigens by these nascent receptors leads to their purging, either by the apoptotic death of the lymphocyte or by the initiation of receptor editing, a process in which the autoreactive receptor is replaced. If the lymphocytes mature further, the activation threshold of autoreactive cells can be tuned by the co-expression of inhibitory receptors or negative signaling molecules, allowing the persistence of the receptor without an increased risk of autoimmunity. T-cell and B-cell receptors that escape these checkpoints can still be regulated in the peripheral immune system by both purging and tuning mechanisms. A separate set of mechanisms, mediated by various regulatory cells, also operates to tune peripheral receptors in a cell-extrinsic fashion. The combined action of these processes ensures that the organism does not suffer autoimmune pathology, even if autoreactive receptors are generated and maintained in the immune system.


Subject(s)
Autoimmunity/physiology , Immune Tolerance/physiology , Lymphocytes/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Autoreceptors/immunology , B-Lymphocytes/immunology , Cell Lineage/immunology , Clonal Anergy/physiology , Humans , Lymphocyte Activation/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...