Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Cell Metab ; 28(6): 946-960.e6, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30078552

ABSTRACT

Although CD8+ T-cell-mediated autoimmune ß cell destruction occurs in type 1 diabetes (T1D), the target epitopes processed and presented by ß cells are unknown. To identify them, we combined peptidomics and transcriptomics strategies. Inflammatory cytokines increased peptide presentation in vitro, paralleling upregulation of human leukocyte antigen (HLA) class I expression. Peptide sources featured several insulin granule proteins and all known ß cell antigens, barring islet-specific glucose-6-phosphatase catalytic subunit-related protein. Preproinsulin yielded HLA-A2-restricted epitopes previously described. Secretogranin V and its mRNA splice isoform SCG5-009, proconvertase-2, urocortin-3, the insulin gene enhancer protein ISL-1, and an islet amyloid polypeptide transpeptidation product emerged as antigens processed into HLA-A2-restricted epitopes, which, as those already described, were recognized by circulating naive CD8+ T cells in T1D and healthy donors and by pancreas-infiltrating cells in T1D donors. This peptidome opens new avenues to understand antigen processing by ß cells and for the development of T cell biomarkers and tolerogenic vaccination strategies.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , Transcriptome/immunology , Animals , Biomarkers/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cell Line , Corticotropin-Releasing Hormone/metabolism , Cytokines/metabolism , HLA Antigens/metabolism , Humans , Insulin/metabolism , Islet Amyloid Polypeptide/metabolism , Mice , Neuroendocrine Secretory Protein 7B2/metabolism , Proprotein Convertase 2/metabolism , Protein Precursors/metabolism , Proteomics/methods , Urocortins/metabolism
2.
Nature ; 559(7715): 627-631, 2018 07.
Article in English | MEDLINE | ID: mdl-30022164

ABSTRACT

The thymus is responsible for generating a diverse yet self-tolerant pool of T cells1. Although the thymic medulla consists mostly of developing and mature AIRE+ epithelial cells, recent evidence has suggested that there is far greater heterogeneity among medullary thymic epithelial cells than was previously thought2. Here we describe in detail an epithelial subset that is remarkably similar to peripheral tuft cells that are found at mucosal barriers3. Similar to the periphery, thymic tuft cells express the canonical taste transduction pathway and IL-25. However, they are unique in their spatial association with cornified aggregates, ability to present antigens and expression of a broad diversity of taste receptors. Some thymic tuft cells pass through an Aire-expressing stage and depend on a known AIRE-binding partner, HIPK2, for their development. Notably, the taste chemosensory protein TRPM5 is required for their thymic function through which they support the development and polarization of thymic invariant natural killer T cells and act to establish a medullary microenvironment that is enriched in the type 2 cytokine, IL-4. These findings indicate that there is a compartmentalized medullary environment in which differentiation of a minor and highly specialized epithelial subset has a non-redundant role in shaping thymic function.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Interleukin-4/metabolism , Thymocytes/cytology , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Cellular Microenvironment , Doublecortin-Like Kinases , Female , Humans , Immune Tolerance/immunology , Interleukin-4/biosynthesis , Interleukins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , Thymocytes/metabolism , Thymus Gland/anatomy & histology , Transcription Factors/deficiency , Transcription Factors/genetics , AIRE Protein
3.
J Immunol Methods ; 457: 82-86, 2018 06.
Article in English | MEDLINE | ID: mdl-29605230

ABSTRACT

The study of organogenesis, tissue-homeostasis and regeneration requires the precise assessment of in vivo cell proliferation. To this end a host of methods have been developed to detect and quantify DNA synthesis in proliferating cells. These include cell labeling with various nucleotide analogues and fluorescence reporter-based animal models with each method presenting its idiosyncratic shortcomings. Quantitative assessment of epithelial cell turnover has been partly hampered due to their variable and limited in vivo accessibility and the requirement for harsher isolation procedures to procure single cells for FACS analysis. Here, we report a reliable protocol to study in vivo cell proliferation of epithelial cells in mice by repeatedly injecting EdU intravenously for an extended 12-day period. EdU incorporation was quantitated ex vivo by FACS after tissue dissociation in order to obtain single epithelial cell suspensions. As a lead population, we analyzed thymic epithelial cells (TECs), where we were able to label compartmentalized TEC subsets to saturation without apparent toxic effects on the thymus architecture or stress-sensitive TEC lineage differentiation. The data is in concordance with the prevailing model of medullary TEC terminal differentiation that includes the post-Aire stage. The same protocol was successfully applied to epithelial cells of various other organs - skin, lymph node, kidney and small intestine - tissues with widely varying frequencies and rates of proliferating epithelial cells.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelial Cells/cytology , Staining and Labeling/methods , Administration, Intravenous , Animals , Cell Count , Deoxyuridine/administration & dosage , Deoxyuridine/analogs & derivatives , Flow Cytometry , Mice , Mice, Inbred C57BL , Thymus Gland/cytology
4.
Sci Immunol ; 3(20)2018 02 02.
Article in English | MEDLINE | ID: mdl-29429978

ABSTRACT

The human leukocyte antigen-A2 (HLA-A2)-restricted zinc transporter 8186-194 (ZnT8186-194) and other islet epitopes elicit interferon-γ secretion by CD8+ T cells preferentially in type 1 diabetes (T1D) patients compared with controls. We show that clonal ZnT8186-194-reactive CD8+ T cells express private T cell receptors and display equivalent functional properties in T1D and healthy individuals. Ex vivo analyses further revealed that CD8+ T cells reactive to ZnT8186-194 and other islet epitopes circulate at similar frequencies and exhibit a predominantly naïve phenotype in age-matched T1D and healthy donors. Higher frequencies of ZnT8186-194-reactive CD8+ T cells with a more antigen-experienced phenotype were detected in children versus adults, irrespective of disease status. Moreover, some ZnT8186-194-reactive CD8+ T cell clonotypes were found to cross-recognize a Bacteroides stercoris mimotope. Whereas ZnT8 was poorly expressed in thymic medullary epithelial cells, variable thymic expression levels of islet antigens did not modulate the peripheral frequency of their cognate CD8+ T cells. In contrast, ZnT8186-194-reactive cells were enriched in the pancreata of T1D patients versus nondiabetic and type 2 diabetic individuals. Thus, islet-reactive CD8+ T cells circulate in most individuals but home to the pancreas preferentially in T1D patients. We conclude that the activation of this common islet-reactive T cell repertoire and progression to T1D likely require defective peripheral immunoregulation and/or a proinflammatory islet microenvironment.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Pancreas/cytology , Pancreas/immunology , Adult , Cell Line , Child , Female , HLA-A2 Antigen/immunology , Healthy Volunteers , Humans , Male
5.
Nat Commun ; 9(1): 353, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367624

ABSTRACT

Establishing effective central tolerance requires the promiscuous expression of tissue-restricted antigens by medullary thymic epithelial cells. However, whether central tolerance also extends to post-translationally modified proteins is not clear. Here we show a mouse model of autoimmunity in which disease development is dependent on post-translational modification (PTM) of the tissue-restricted self-antigen collagen type II. T cells specific for the non-modified antigen undergo efficient central tolerance. By contrast, PTM-reactive T cells escape thymic selection, though the PTM variant constitutes the dominant form in the periphery. This finding implies that the PTM protein is absent in the thymus, or present at concentrations insufficient to induce negative selection of developing thymocytes and explains the lower level of tolerance induction against the PTM antigen. As the majority of self-antigens are post-translationally modified, these data raise the possibility that T cells specific for other self-antigens naturally subjected to PTM may escape central tolerance induction by a similar mechanism.


Subject(s)
Arthritis, Experimental/immunology , Central Tolerance/immunology , Collagen Type II/immunology , Protein Processing, Post-Translational/immunology , T-Lymphocytes/immunology , Animals , Autoantigens/immunology , Autoimmunity/immunology , Disease Models, Animal , Mice , Mice, Transgenic , Thymocytes/immunology , Thymus Gland/immunology
6.
Front Immunol ; 8: 1511, 2017.
Article in English | MEDLINE | ID: mdl-29170668

ABSTRACT

Immunization with myelin components can elicit experimental autoimmune encephalomyelitis (EAE). EAE susceptibility varies between mouse strains, depending on the antigen employed. BL/6 mice are largely resistant to EAE induction with proteolipid protein (PLP), probably a reflection of antigen-specific tolerance. However, the extent and mechanism(s) of tolerance to PLP remain unclear. Here, we identified three PLP epitopes in PLP-deficient BL/6 mice. PLP-sufficient mice did not respond against two of these, whereas tolerance was "leaky" for an epitope with weak predicted MHCII binding, and only this epitope was encephalitogenic. In TCR transgenic mice, the "EAE-susceptibility-associated" epitope was "ignored" by specific CD4 T cells, whereas the "resistance-associated" epitope induced clonal deletion and Treg induction in the thymus. Central tolerance was autoimmune regulator dependent and required expression and presentation of PLP by thymic epithelial cells (TECs). TEC-specific ablation of PLP revealed that peripheral tolerance, mediated by dendritic cells through recessive tolerance mechanisms (deletion and anergy), could largely compensate for a lack of central tolerance. However, adoptive EAE was exacerbated in mice lacking PLP in TECs, pointing toward a non-redundant role of the thymus in dominant tolerance to PLP. Our findings reveal multiple layers of tolerance to a central nervous system autoantigen that vary among epitopes and thereby specify disease susceptibility. Understanding how different modalities of tolerance apply to distinct T cell epitopes of a target in autoimmunity has implications for antigen-specific strategies to therapeutically interfere with unwanted immune reactions against self.

7.
J Immunol ; 199(10): 3488-3503, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28993517

ABSTRACT

The basic two-step terminal differentiation model of the medullary thymic epithelial cell (mTEC) lineage from immature MHC class II (MHCII)lo to mature MHCIIhi mTECs has recently been extended to include a third stage, namely the post-Aire MHCIIlo subset as identified by lineage-tracing models. However, a suitable surface marker distinguishing the phenotypically overlapping pre- from the post-Aire MHCIIlo stage has been lacking. In this study, we introduce the lectin Tetragonolobus purpureas agglutinin (TPA) as a novel cell surface marker that allows for such delineation. Based on our data, we derived the following sequence of mTEC differentiation: TPAloMHCIIlo → TPAloMHCIIhi → TPAhiMHCIIhi → TPAhiMHCIIlo Surprisingly, in the steady-state postnatal thymus TPAloMHCIIlo pre-Aire rather than terminally differentiated post-Aire TPAhiMHCIIlo mTECs were marked for apoptosis at an exceptionally high rate of ∼70%. Hence, only the minor cycling fraction of the MHCIIlo subset (<20%) potentially qualified as mTEC precursors. FoxN1 expression inversely correlated with the fraction of slow cycling and apoptotic cells within the four TPA subsets. TPA also further subdivided human mTECs, although with different subset distribution. Our revised road map emphazises close parallels of terminal mTEC development with that of skin, undergoing an alternative route of cell death, namely cornification rather than apoptosis. The high rate of apoptosis in pre-Aire MHCIIlo mTECs points to a "quality control" step during early mTEC differentiation.


Subject(s)
Biomarkers/metabolism , Epithelial Cells/physiology , Lectins/metabolism , Thymus Gland/cytology , Transcription Factors/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Lineage , Cells, Cultured , Female , Gene Expression Regulation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Rats , Rats, Sprague-Dawley , Thymus Gland/anatomy & histology , Transcription Factors/genetics , AIRE Protein
8.
Eur J Immunol ; 47(7): 1153-1159, 2017 07.
Article in English | MEDLINE | ID: mdl-28439878

ABSTRACT

The origin of the thymic epithelium, i.e. the cortical (cTEC) and medullary (mTEC) epithelial cells, from bipotent stem cells through TEC progenitors and lineage-specific progeny still remains poorly understood. We sought to obtain an unbiased view of the incipient emergence of TEC subsets by following embryonic TEC development based on co-expression of EpCAM, CD80 and MHC class II (MHCII) on non-hematopoietic (CD45- ) thymic stromal cells in wild-type BL6 mice. Using a combination of ex vivo analysis, Re-aggregate Thymic Organ Culture (RTOC) reconstitution assays and mathematical modeling, we traced emergent lineage commitment in murine embryonic TECs. Both experimental and mathematical datasets supported the following developmental sequence: MHCII- CD80- → MHCIIlo CD80- → MHCIIhi CD80- → MHCIIhi CD80hi TECs, whereby MHCIIhi CD80- and MHCIIhi CD80hi TECs bear features of cTECs and mTECs respectively. These emergent MHCIIhi CD80- cTECs directly generate mature MHCIIhi CD80hi mTECs in vivo and in vitro, thus supporting the asynchronous model of TEC lineage commitment.


Subject(s)
Cell Differentiation , Epithelial Cells/physiology , Thymocytes/physiology , Thymus Gland/cytology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Cell Lineage , Cells, Cultured , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/immunology , Epithelial Cells/immunology , Gene Expression , Genes, MHC Class II/genetics , Genes, MHC Class II/immunology , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Mice , Models, Theoretical , Organ Culture Techniques , Thymus Gland/embryology , Thymus Gland/immunology
9.
Cell Rep ; 17(2): 448-457, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705793

ABSTRACT

Adult tissue-specific stem cells (SCs) mediate tissue homeostasis and regeneration and can give rise to all lineages in the corresponding tissue, similar to the early progenitors that generate organs in the first place. However, the developmental origins of adult SCs are largely unknown. We recently identified thymosphere-forming stem cells (TSFCs) in the adult mouse thymus, which display genuine stemness features and can generate the two major thymic epithelial cell lineages. Here, we show that embryonic TSFCs possess stemness features but differ from adult TSFCs in surface marker profile. Our findings support the model of a continuous thymic SC lineage that is maintained throughout ontogeny. TGF-ß signaling differentially affects embryonic versus adult thymosphere formation, suggesting that thymic epithelial SC potency depends on both developmental stage and environmental signals. Collectively, our findings suggest that embryonic TSFCs contribute to an adult SC pool and that TSFC plasticity is controlled by TGF-ß signaling.


Subject(s)
Embryonic Stem Cells/cytology , Epithelial Cells/cytology , Thymus Gland/cytology , Transforming Growth Factor beta/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Embryonic Stem Cells/metabolism , Epithelial Cells/metabolism , Mice , Regeneration/genetics , Signal Transduction , Thymus Gland/growth & development
10.
Eur J Immunol ; 46(9): 2247-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27334749

ABSTRACT

T-cell polyspecificity, predicting that individual T cells recognize a continuum of related ligands, implies that multiple antigens can tolerize T cells specific for a given self-antigen. We previously showed in C57BL/6 mice that part of the CD4(+) T-cell repertoire specific for myelin oligodendrocyte glycoprotein (MOG) 35-55 also recognizes the neuronal antigen neurofilament medium (NF-M) 15-35. Such bi-specific CD4(+) T cells are frequent and produce inflammatory cytokines after stimulation. Since T cells recognizing two self-antigens would be expected to be tolerized more efficiently, this finding prompted us to study how polyspecificity impacts tolerance. We found that similar to MOG, NF-M is expressed in the thymus by medullary thymic epithelial cells, a tolerogenic population. Nevertheless, the frequency, phenotype, and capacity to transfer experimental autoimmune encephalomyelitis (EAE) of MOG35-55 -reactive CD4(+) T cells were increased in MOG-deficient but not in NF-M-deficient mice. We found that presentation of NF-M15-35 by I-A(b) on dendritic cells is of short duration, suggesting unstable MHC class II binding. Consistently, introducing an MHC-anchoring residue into NF-M15-35 (NF-M15-35 T20Y) increased its immunogenicity, activating a repertoire able to induce EAE. Our results show that in C57BL/6 mice bi-specific encephalitogenic T cells manage to escape tolerization due to inefficient exposure to two self-antigens.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immune Tolerance , Myelin Proteins/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurons/immunology , Animals , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/genetics , Neurofilament Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Cell Antigen Receptor Specificity/genetics , T-Cell Antigen Receptor Specificity/immunology , Thymus Gland/immunology , Thymus Gland/metabolism
12.
Trends Immunol ; 37(2): 114-125, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26795134

ABSTRACT

Recent studies have leveraged MHC tetramer and TCR sequencing approaches towards a more precise characterization of the peripheral T cell repertoire, providing important insight into both the contribution of self-reactive T cells to the overall repertoire and their function. The peripheral T cell repertoire of healthy individuals contains a high frequency of diverse, self-reactive T cells. Furthermore, self-reactive T cells can perform essential beneficial physiological functions. We review these recent findings here, and discuss their implications to the current understanding of peripheral tolerance and the role of self-reactive T cells in autoimmune disease. We outline gaps in understanding, and argue that an important step forward is to revise the definition of self-reactive T cells to incorporate new concepts regarding the nature and physiological functions of different populations of T cells capable of recognizing self-antigens.


Subject(s)
Autoimmune Diseases/immunology , T-Lymphocyte Subsets/physiology , T-Lymphocytes/physiology , Animals , Autoantigens/immunology , Humans , Peripheral Tolerance , Receptors, Antigen, T-Cell/metabolism
13.
J Autoimmun ; 67: 65-75, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26481130

ABSTRACT

Promiscuous expression of a plethora of tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs) is essential for central tolerance. This promiscuous gene expression (pGE) is characterized by inclusion of a broad range of TRAs and by its mosaic expression patterns, i.e. each antigen is only expressed in 1-3% of mTECs. It is currently unclear to which extent random and/or deterministic mechanisms are involved in the regulation of pGE. In order to address this issue, we deconstructed the transcriptional heterogeneity in mTEC to minor subsets expressing a particular TRA. We identified six delineable co-expression groups in mouse mTECs. These co-expression groups displayed a variable degree of mutual overlap and mapped to different stages of mTEC development. Co-expressed genes showed chromosomal preference and clustered within delimited genomic regions. Moreover, co-expression groups in mice and humans selected by a pair of orthologous genes preferentially co-expressed sets of orthologous genes attesting to the species conservation of pGE between mouse and human. Furthermore, co-expressed genes were enriched for specific transcription factor binding motifs concomitant with up-regulation of the corresponding transcription factors, implicating additional factors in the regulation of pGE besides the Autoimmune Regulator (Aire). Thus promiscuous transcription of self-antigens in mTECs entails a highly coordinated process, which is evolutionary strictly conserved between species.


Subject(s)
Autoantigens/genetics , Autoantigens/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression Regulation , Genetic Variation , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , Binding Sites , Cell Differentiation/genetics , Cell Differentiation/immunology , Cluster Analysis , Epithelial Cells/cytology , Evolution, Molecular , Gene Expression Profiling , Humans , Mice , Mice, Transgenic , Nucleotide Motifs , Organ Specificity/genetics , Organ Specificity/immunology , Promoter Regions, Genetic , Protein Binding , Thymus Gland/cytology , Transcription Factors/metabolism , Transcriptional Activation
14.
Genom Data ; 6: 48-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26697330

ABSTRACT

The establishment of central tolerance essentially depends on the promiscuous gene expression (pGE) of a plethora of tissue restricted antigens by the medullary thymic epithelial cells. The antigens are presented to developing thymocytes in the thymus to select for non-self reactive T-cell receptors in order to prevent autoimmune reactions in the periphery. However the molecular regulation of tissue-restricted antigen expression is still poorly understood. The only regulator known to play a role in the transcriptional regulation so far is the autoimmune regulator (AIRE). AIRE is thought to act in a multi-protein complex, promoting transcription, elongation and splicing of target genes. Yet the full composition of this Aire-associated multi-protein complex and its mode of action remain to be elucidated. Here we describe the experimental details and controls of the gene array analysis on the impact of the homeodomain-interacting protein kinase 2 (Hipk2) on promiscuous gene expression in medullary thymic epithelial cells based on the analysis of newly generated TEC-specific Hipk2 conditional knockout mice. The changes in gene expression are presumably mediated through a regulatory effect of Hipk2 on AIRE as published in the study by Rattay and colleagues in the Journal of Immunology [1]. The gene array data reported in this paper have been deposited in the Gene Expression Omnibus (GEO) database, www.ncbi.nlm.nih.gov/geo (accession no. GSE63432).

15.
J Immunol ; 195(11): 5261-71, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26512137

ABSTRACT

Wnt signaling has been implicated in T cell development. However, it remained unclear which cell type is the major source of Wnt ligands and to what extent thymic epithelial cell (TEC) development is dependent on Wnt signaling. In this study, we analyzed the role of Wnt ligands provided by TECs for the development of T cells and TECs without manipulating the intracellular Wnt signaling machinery in either cell type. To this end, we used conditional knockout mice (FoxN1-Gpr177) in which TECs are unable to secrete Wnt ligands. Gpr177 (Evi/Wls) is a Wnt-specific cargo receptor that is required for the secretion of Wnt ligands. We found that TECs are the main source of Wnt ligands in the thymus, which serves a nonredundant role, and lack of TEC-provided Wnt ligands led to thymic hypotrophy, as well as a reduced peripheral T cell pool. Despite being reduced in numbers, T cells that developed in the absence of TEC-secreted Wnt ligands were functionally competent, and the subset composition of the peripheral T cell pool was not affected. Thus, our data suggest that T cell development is not directly dependent on TEC-provided Wnt ligands. Rather, TEC-secreted Wnt ligands are essential for normal thymus development and normal peripheral T cell frequencies but are dispensable for T cell function in the periphery.


Subject(s)
Lymphocyte Activation/immunology , T-Lymphocytes/cytology , Thymus Gland/immunology , Wnt Proteins/metabolism , Animals , Cell Differentiation/immunology , Epithelial Cells/immunology , Forkhead Transcription Factors/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , T-Lymphocytes/immunology , Wnt Signaling Pathway/immunology
16.
J Vis Exp ; (101): e52614, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26275017

ABSTRACT

Intra-thymic T cell development requires an intricate three-dimensional meshwork composed of various stromal cells, i.e., non-T cells. Thymocytes traverse this scaffold in a highly coordinated temporal and spatial order while sequentially passing obligatory check points, i.e., T cell lineage commitment, followed by T cell receptor repertoire generation and selection prior to their export into the periphery. The two major resident cell types forming this scaffold are cortical (cTECs) and medullary thymic epithelial cells (mTECs). A key feature of mTECs is the so-called promiscuous expression of numerous tissue-restricted antigens. These tissue-restricted antigens are presented to immature thymocytes directly or indirectly by mTECs or thymic dendritic cells, respectively resulting in self-tolerance. Suitable in vitro models emulating the developmental pathways and functions of cTECs and mTECs are currently lacking. This lack of adequate experimental models has for instance hampered the analysis of promiscuous gene expression, which is still poorly understood at the cellular and molecular level. We adapted a 3D organotypic co-culture model to culture ex vivo isolated mTECs. This model was originally devised to cultivate keratinocytes in such a way as to generate a skin equivalent in vitro. The 3D model preserved key functional features of mTEC biology: (i) proliferation and terminal differentiation of CD80(lo), Aire-negative into CD80(hi), Aire-positive mTECs, (ii) responsiveness to RANKL, and (iii) sustained expression of FoxN1, Aire and tissue-restricted genes in CD80(hi) mTECs.


Subject(s)
Coculture Techniques/methods , Epithelial Cells/physiology , Thymocytes/physiology , Thymus Gland/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Epithelial Cells/cytology , Female , Gene Expression , Mice , Mice, Inbred C57BL , Pregnancy , RANK Ligand , Stromal Cells/cytology , Thymocytes/cytology , Thymus Gland/cytology
17.
Nat Immunol ; 16(9): 933-41, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26237553

ABSTRACT

Expression of tissue-restricted self antigens (TRAs) in medullary thymic epithelial cells (mTECs) is essential for the induction of self-tolerance and prevents autoimmunity, with each TRA being expressed in only a few mTECs. How this process is regulated in single mTECs and is coordinated at the population level, such that the varied single-cell patterns add up to faithfully represent TRAs, is poorly understood. Here we used single-cell RNA sequencing and obtained evidence of numerous recurring TRA-co-expression patterns, each present in only a subset of mTECs. Co-expressed genes clustered in the genome and showed enhanced chromatin accessibility. Our findings characterize TRA expression in mTECs as a coordinated process that might involve local remodeling of chromatin and thus ensures a comprehensive representation of the immunological self.


Subject(s)
Autoantigens/genetics , Epithelial Cells/immunology , Gene Expression Regulation/immunology , RNA, Messenger/metabolism , Self Tolerance/immunology , Thymus Gland/immunology , Animals , Autoimmunity/immunology , Chromatin Assembly and Disassembly , Epithelial Cells/metabolism , Gene Expression Profiling , Humans , Mice , Self Tolerance/genetics , Single-Cell Analysis , Thymus Gland/cytology , Thymus Gland/metabolism
18.
Immunity ; 42(6): 1048-61, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26070482

ABSTRACT

Thymic antigen-presenting cells (APCs) such as dendritic cells and medullary thymic epithelial cells (mTECs) use distinct strategies of self-antigen expression and presentation to mediate central tolerance. The thymus also harbors B cells; whether they also display unique tolerogenic features and how they genealogically relate to peripheral B cells is unclear. Here, we found that Aire is expressed in thymic but not peripheral B cells. Aire expression in thymic B cells coincided with major histocompatibility class II (MHCII) and CD80 upregulation and immunoglobulin class-switching. These features were recapitulated upon immigration of naive peripheral B cells into the thymus, whereby this intrathymic licensing required CD40 signaling in the context of cognate interactions with autoreactive CD4(+) thymocytes. Moreover, a licensing-dependent neo-antigen selectively upregulated in immigrating B cells mediated negative selection through direct presentation. Thus, autoreactivity within the nascent T cell repertoire fuels a feed forward loop that endows thymic B cells with tolerogenic features.


Subject(s)
B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/metabolism , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Antigen Presentation/genetics , Autoantigens/immunology , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , CD40 Antigens/genetics , Cell Differentiation/genetics , Cells, Cultured , Central Tolerance/genetics , Clonal Selection, Antigen-Mediated/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Immunoglobulin Class Switching/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction , Transcription Factors/genetics , AIRE Protein
19.
J Autoimmun ; 60: 12-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25911201

ABSTRACT

Promiscuous gene expression (pGE) of tissue-restricted self-antigens (TRA) in medullary thymic epithelial cells (mTECs) is in part driven by the Autoimmune Regulator gene (AIRE) and essential for self-tolerance. The link between AIRE functional mutations and multi-organ autoimmunity in human and mouse supports the role of pGE. Deep sequencing of the transcriptome revealed that mouse mTECs potentially transcribe an unprecedented range of >90% of all genes. Yet, it remains unclear to which extent these low-level transcripts are actually translated into proteins, processed and presented by thymic APCs to induce tolerance. To address this, we analyzed the HLA-DR-associated thymus peptidome. Within a large panel of peptides from abundant proteins, two TRA peptides were identified: prostate-specific semenogelin-1 (an autoantigen in autoimmune chronic prostatitis/chronic pelvic pain syndrome) and central nervous system-specific contactin-2 (an autoantigen in multiple sclerosis). Thymus expression of both genes was restricted to mTECs. SEMG1 expression was confined to mature HLA-DR(hi) mTECs of male and female donors and was AIRE-dependent, whereas CNTN2 was apparently AIRE-independent and was expressed by both populations of mTECs. Our findings establish a link between pGE, MHC-II peptide presentation and autoimmunity for bona fide human TRAs.


Subject(s)
Autoantigens/immunology , HLA-DR Antigens/immunology , Self Tolerance/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Autoantigens/biosynthesis , Autoimmunity/immunology , Child , Child, Preschool , Contactin 2/biosynthesis , Contactin 2/immunology , Epithelial Cells/immunology , Female , Gene Expression Profiling , Humans , Infant , Infant, Newborn , Male , Mice , Middle Aged , Seminal Vesicle Secretory Proteins/biosynthesis , Seminal Vesicle Secretory Proteins/immunology , Thymus Gland/cytology , Transcription Factors/biosynthesis , Transcriptome , Young Adult , AIRE Protein
20.
Diabetes ; 64(10): 3532-42, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25918233

ABSTRACT

The first signs of autoimmune activation leading to ß-cell destruction in type 1 diabetes (T1D) appear during the first months of life. Thus, the perinatal period offers a suitable time window for disease prevention. Moreover, thymic selection of autoreactive T cells is most active during this period, providing a therapeutic opportunity not exploited to date. We therefore devised a strategy by which the T1D-triggering antigen preproinsulin fused with the immunoglobulin (Ig)G Fc fragment (PPI-Fc) is delivered to fetuses through the neonatal Fc receptor (FcRn) pathway, which physiologically transfers maternal IgGs through the placenta. PPI-Fc administered to pregnant PPIB15-23 T-cell receptor-transgenic mice efficiently accumulated in fetuses through the placental FcRn and protected them from subsequent diabetes development. Protection relied on ferrying of PPI-Fc to the thymus by migratory dendritic cells and resulted in a rise in thymic-derived CD4(+) regulatory T cells expressing transforming growth factor-ß and in increased effector CD8(+) T cells displaying impaired cytotoxicity. Moreover, polyclonal splenocytes from nonobese diabetic (NOD) mice transplacentally treated with PPI-Fc were less diabetogenic upon transfer into NOD.scid recipients. Transplacental antigen vaccination provides a novel strategy for early T1D prevention and, further, is applicable to other immune-mediated conditions.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Histocompatibility Antigens Class I/metabolism , Insulin/metabolism , Maternal-Fetal Exchange/physiology , Protein Precursors/metabolism , Receptors, Fc/metabolism , Animals , Autoimmunity , Cell Proliferation , Dendritic Cells/physiology , Female , Gene Expression Regulation, Developmental/physiology , Histocompatibility Antigens Class I/genetics , Humans , Insulin/administration & dosage , Mice , Mice, Inbred NOD , Mice, Transgenic , Placenta/metabolism , Pregnancy , Protein Precursors/administration & dosage , Receptors, Fc/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Thymus Gland/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...