Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 16(4): 373-385, 2023 08.
Article in English | MEDLINE | ID: mdl-36739089

ABSTRACT

Interleukin (IL)-17 protects epithelial barriers by inducing the secretion of antimicrobial peptides. However, the effect of IL-17 on Paneth cells (PCs), the major producers of antimicrobial peptides in the small intestine, is unclear. Here, we show that the targeted ablation of the IL-17 receptor (IL-17R) in PCs disrupts their antimicrobial functions and decreases the frequency of ileal PCs. These changes become more pronounced after colonization with IL-17 inducing segmented filamentous bacteria. Mice with PCs that lack IL-17R show an increased inflammatory transcriptional profile in the ileum along with the severity of experimentally induced ileitis. These changes are associated with a decrease in the diversity of gut microbiota that induces a severe ileum pathology upon transfer to genetically susceptible mice, which can be prevented by the systemic administration of IL-17a/f in microbiota recipients. In an exploratory analysis of a small cohort of pediatric patients with Crohn's disease, we have found that a portion of these patients exhibits a low number of lysozyme-expressing ileal PCs and a high ileitis severity score, resembling the phenotype of mice with IL-17R-deficient PCs. Our study identifies IL-17R-dependent signaling in PCs as an important mechanism that maintains ileal homeostasis through the prevention of dysbiosis.


Subject(s)
Ileitis , Microbiota , Receptors, Interleukin-17 , Animals , Child , Humans , Mice , Antimicrobial Peptides , Dysbiosis/microbiology , Ileitis/microbiology , Ileum/microbiology , Inflammation/pathology , Interleukin-17 , Paneth Cells/pathology , Receptors, Interleukin-17/genetics
2.
Proc Natl Acad Sci U S A ; 120(9): e2220120120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36802427

ABSTRACT

The activation of thymic B cells is critical for their licensing as antigen presenting cells and resulting ability to mediate T cell central tolerance. The processes leading to licensing are still not fully understood. By comparing thymic B cells to activated Peyer's patch B cells at steady state, we found that thymic B cell activation starts during the neonatal period and is characterized by TCR/CD40-dependent activation, followed by immunoglobulin class switch recombination (CSR) without forming germinal centers. Transcriptional analysis also demonstrated a strong interferon signature, which was not apparent in the periphery. Thymic B cell activation and CSR were primarily dependent on type III IFN signaling, and loss of type III IFN receptor in thymic B cells resulted in reduced thymocyte regulatory T cell (Treg) development. Finally, from TCR deep sequencing, we estimate that licensed B cells induce development of a substantial fraction of the Treg cell repertoire. Together, these findings reveal the importance of steady-state type III IFN in generating licensed thymic B cells that induce T cell tolerance to activated B cells.


Subject(s)
Interferon Lambda , T-Lymphocytes, Regulatory , Humans , Infant, Newborn , Thymus Gland , Thymocytes , Receptors, Antigen, T-Cell
3.
Front Immunol ; 13: 926625, 2022.
Article in English | MEDLINE | ID: mdl-35774801

ABSTRACT

The inevitability of evolution of the adaptive immune system with its mechanism of randomly rearranging segments of the T cell receptor (TCR) gene is the generation of self-reactive clones. For the sake of prevention of autoimmunity, these clones must be eliminated from the pool of circulating T cells. This process occurs largely in the thymic medulla where the strength of affinity between TCR and self-peptide MHC complexes is the factor determining thymocyte fate. Thus, the display of self-antigens in the thymus by thymic antigen presenting cells, which are comprised of medullary thymic epithelial (mTECs) and dendritic cells (DCs), is fundamental for the establishment of T cell central tolerance. Whereas mTECs produce and present antigens in a direct, self-autonomous manner, thymic DCs can acquire these mTEC-derived antigens by cooperative antigen transfer (CAT), and thus present them indirectly. While the basic characteristics for both direct and indirect presentation of self-antigens are currently known, recent reports that describe the heterogeneity of mTEC and DC subsets, their presentation capacity, and the potentially non-redundant roles in T cell selection processes represents another level of complexity which we are attempting to unravel. In this review, we underscore the seminal studies relevant to these topics with an emphasis on new observations pertinent to the mechanism of CAT and its cellular trajectories underpinning the preferential distribution of thymic epithelial cell-derived self-antigens to specific subsets of DC. Identification of molecular determinants which control CAT would significantly advance our understanding of how the cellularly targeted presentation of thymic self-antigens is functionally coupled to the T cell selection process.


Subject(s)
Autoantigens , Dendritic Cells , Central Tolerance , Epithelial Cells , Receptors, Antigen, T-Cell , Thymus Gland
4.
Nat Immunol ; 23(7): 1042-1051, 2022 07.
Article in English | MEDLINE | ID: mdl-35637352

ABSTRACT

The thymus contains a diversity of dendritic cells (DCs) that exist in defined locations and have different antigen-processing and -presenting features. This suggests that they play nonredundant roles in mediating thymocyte selection. In an effort to eliminate SIRPα+ classic DC2 subsets, we discovered that a substantial proportion expresses the surface lectin, CD301b, in the thymus. These cells resemble the CD301b+ type 2 immune response promoting DCs that are present in the skin-draining lymph nodes. Transcriptional and phenotypic comparison to other DC subsets in the thymus revealed that thymic CD301b+ cDCs represent an activated state that exhibits enhanced antigen processing and presentation. Furthermore, a CD301b+ cDC2 subset demonstrated a type 2 cytokine signature and required steady-state interleukin-4 receptor signaling. Selective ablation of CD301b+ cDC2 subsets impaired clonal deletion without affecting regulatory T cells (Treg cells). The T cell receptor α repertoire sequencing confirmed that a cDC2 subset promotes deletion of conventional T cells with minimal effect on Treg cell selection. Together, these findings suggest that cytokine-induced activation of DCs in the thymus substantially enforces central tolerance.


Subject(s)
Clonal Deletion , Dendritic Cells , Antigen Presentation , Cytokines , Lymphocyte Activation , Thymus Gland
5.
Elife ; 112022 01 31.
Article in English | MEDLINE | ID: mdl-35099391

ABSTRACT

Medullary thymic epithelial cells (mTECs), which produce and present self-antigens, are essential for the establishment of central tolerance. Since mTEC numbers are limited, their function is complemented by thymic dendritic cells (DCs), which transfer mTEC-produced self-antigens via cooperative antigen transfer (CAT). While CAT is required for effective T cell selection, many aspects remain enigmatic. Given the recently described heterogeneity of mTECs and DCs, it is unclear whether the antigen acquisition from a particular TEC subset is mediated by preferential pairing with a specific subset of DCs. Using several relevant Cre-based mouse models that control for the expression of fluorescent proteins, we have found that, in regards to CAT, each subset of thymic DCs preferentially targets a distinct mTEC subset(s). Importantly, XCR1+-activated DC subset represented the most potent subset in CAT. Interestingly, thymic DCs can also acquire antigens from more than one mTEC, and of these, monocyte-derived dendritic cells (moDCs) were determined to be the most efficient. moDCs also represented the most potent DC subset in the acquisition of antigen from other DCs. These findings suggest a preferential pairing model for the distribution of mTEC-derived antigens among distinct populations of thymic DCs.


Subject(s)
Antigen Presentation/immunology , Autoantigens/metabolism , Immune Tolerance , Thymus Gland/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Thymus Gland/cytology
6.
Eur J Immunol ; 51(9): 2237-2250, 2021 09.
Article in English | MEDLINE | ID: mdl-34107067

ABSTRACT

Early embryonic hematopoiesis in mammals is defined by three successive waves of hematopoietic progenitors which exhibit a distinct hematopoietic potential and provide continuous support for the development of the embryo and adult organism. Although the functional importance of each of these waves has been analyzed, their spatio-temporal overlap and the lack of wave-specific markers hinders the accurate separation and assessment of their functional roles during early embryogenesis. We have recently shown that TLR2, in combination with c-kit, represents the earliest signature of emerging precursors of the second hematopoietic wave, erythro-myeloid precursors (EMPs). Since the onset of Tlr2 expression distinguishes EMPs from primitive progenitors which coexist in the yolk sac from E7.5, we generated a novel transgenic "knock in" mouse model, Tlr2Dtr , suitable for inducible targeted depletion of TLR2+ EMPs. In this model, the red fluorescent protein and diphtheria toxin receptor sequences are linked via a P2A sequence and inserted into the Tlr2 locus before its stop codon. We show that a timely controlled deletion of TLR2+ EMPs in Tlr2Dtr embryos results in a marked decrease in both erythroid as well as myeloid lineages and, consequently, in embryonic lethality peaking before E13.5. These findings validate the importance of EMPs in embryonic development.


Subject(s)
Embryo, Mammalian/pathology , Embryonic Development/genetics , Hematopoiesis/genetics , Myeloid Progenitor Cells/cytology , Toll-Like Receptor 2/genetics , Animals , Embryo, Mammalian/embryology , Erythrocytes/cytology , Hematopoiesis/physiology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
Nat Commun ; 11(1): 2361, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32398640

ABSTRACT

The development of thymic regulatory T cells (Treg) is mediated by Aire-regulated self-antigen presentation on medullary thymic epithelial cells (mTECs) and dendritic cells (DCs), but the cooperation between these cells is still poorly understood. Here we show that signaling through Toll-like receptors (TLR) expressed on mTECs regulates the production of specific chemokines and other genes associated with post-Aire mTEC development. Using single-cell RNA-sequencing, we identify a new thymic CD14+Sirpα+ population of monocyte-derived dendritic cells (CD14+moDC) that are enriched in the thymic medulla and effectively acquire mTEC-derived antigens in response to the above chemokines. Consistently, the cellularity of CD14+moDC is diminished in mice with MyD88-deficient TECs, in which the frequency and functionality of thymic CD25+Foxp3+ Tregs are decreased, leading to aggravated mouse experimental colitis. Thus, our findings describe a TLR-dependent function of mTECs for the recruitment of CD14+moDC, the generation of Tregs, and thereby the establishment of central tolerance.


Subject(s)
Colitis/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Adoptive Transfer , Animals , Antigen Presentation , Autoantigens/immunology , Cell Separation , Chemokines/immunology , Chemokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Flow Cytometry , Lipopolysaccharide Receptors/metabolism , Mice , Receptors, Immunologic/metabolism , Self Tolerance , Sequence Analysis, RNA , Signal Transduction/immunology , Single-Cell Analysis , T-Lymphocytes, Regulatory/transplantation , Thymus Gland/cytology , Toll-Like Receptors/metabolism , Up-Regulation
8.
J Exp Med ; 216(5): 1027-1037, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30918005

ABSTRACT

The autoimmune regulator (Aire) serves an essential function for T cell tolerance by promoting the "promiscuous" expression of tissue antigens in thymic epithelial cells. Aire is also detected in rare cells in peripheral lymphoid organs, but the identity of these cells is poorly understood. Here, we report that Aire protein-expressing cells in lymph nodes exhibit typical group 3 innate lymphoid cell (ILC3) characteristics such as lymphoid morphology, absence of "classical" hematopoietic lineage markers, and dependence on RORγt. Aire+ cells are more frequent among lineage-negative RORγt+ cells of peripheral lymph nodes as compared with mucosa-draining lymph nodes, display a unique Aire-dependent transcriptional signature, express high surface levels of MHCII and costimulatory molecules, and efficiently present an endogenously expressed model antigen to CD4+ T cells. These findings define a novel type of ILC3-like cells with potent APC features, suggesting that these cells serve a function in the control of T cell responses.


Subject(s)
Antigen-Presenting Cells/immunology , Lymph Nodes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , CD11 Antigens/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Gene Expression Regulation , Histocompatibility Antigens Class II/metabolism , Immunity, Innate , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Transcription, Genetic , AIRE Protein
9.
Semin Cell Dev Biol ; 88: 138-146, 2019 04.
Article in English | MEDLINE | ID: mdl-29355606

ABSTRACT

The gut is the biggest immune organ in the body that encloses commensal microbiota which aids in food digestion. Paneth cells, positioned at the frontline of host-microbiota interphase, can modulate the composition of microbiota. Paneth cells achieve this via the delivery of microbicidal substances, among which enteric α-defensins play the primary role. If microbiota is dysregulated, it can impact the function of the local mucosal immune system. Importantly, this system is also exposed to an enormous number of antigens which are derived from the gut-resident microbiota and processed food, and may potentially trigger undesirable local inflammatory responses. To understand the intricate regulations and liaisons between Paneth cells, microbiota and the immune system in this intestinal-specific setting, one must consider their mode of interaction in a wider context of regulatory processes which impose immune tolerance not only to self, but also to microbiota and food-derived antigens. These include, but are not limited to, tolerogenic mechanisms of central tolerance in the thymus and peripheral tolerance in the secondary lymphoid organs, and the intestine itself. Defects in these processes can compromise homeostasis in the intestinal mucosal immunity. In this review, which is focused on tolerance to intestinal antigens and its relevance for the pathogenesis of gut immune diseases, we provide an outline of such multilayered immune control mechanisms and highlight functional links that underpin their cooperative nature.


Subject(s)
Dysbiosis/prevention & control , Gastrointestinal Tract/immunology , Paneth Cells/immunology , Peripheral Tolerance , alpha-Defensins/immunology , Animals , Central Tolerance , Dysbiosis/immunology , Dysbiosis/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Gene Expression/immunology , Homeostasis/immunology , Humans , Immunity, Mucosal/drug effects , Inflammation , Paneth Cells/drug effects , Paneth Cells/microbiology , Symbiosis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/microbiology , alpha-Defensins/biosynthesis , alpha-Defensins/pharmacology
10.
Eur J Immunol ; 48(3): 546-548, 2018 03.
Article in English | MEDLINE | ID: mdl-29193031

ABSTRACT

Medullary thymic epithelial cell (mTEC)-restricted expression of autoimmune regulator (Aire) is essential for establishment of immune tolerance. Recently, Aire was also shown to be expressed in cells of hematopietic and reproductive lineages. Thus, the generation of Airefl/fl mouse strain enables the investigation of the cell-specific function of Aire.


Subject(s)
Immune Tolerance/genetics , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Animals , Antigen-Presenting Cells/pathology , Cell Lineage/genetics , Cell Lineage/immunology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Polyendocrinopathies, Autoimmune/pathology , Reproduction/genetics , Reproduction/immunology , AIRE Protein
11.
Gastroenterology ; 149(1): 139-50, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25982289

ABSTRACT

BACKGROUND & AIMS: Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED) is an autoimmune disorder characterized by chronic mucocutaneous candidiasis, hypoparathyroidism, and adrenal insufficiency, but patients also develop intestinal disorders. APECED is an autosomal recessive disorder caused by mutations in the autoimmune regulator (AIRE, which regulates immune tolerance) that allow self-reactive T cells to enter the periphery. Enteric α-defensins are antimicrobial peptides secreted by Paneth cells. Patients with APECED frequently have gastrointestinal symptoms and seroreactivity against secretory granules of Paneth cells. We investigated whether enteric α-defensins are autoantigens in humans and mice with AIRE deficiency. METHODS: We analyzed clinical data, along with serum and stool samples and available duodenal biopsies from 50 patients with APECED collected from multiple centers in Europe. Samples were assessed for expression of defensins and other molecules by quantitative reverse transcription polymerase chain reaction and flow cytometry; levels of antibodies and other proteins were measured by immunohistochemical and immunoblot analyses. Histologic analyses were performed on biopsy samples. We used Aire(-/-) mice as a model of APECED, and studied the effects of transferring immune cells from these mice to athymic mice. RESULTS: Enteric defensins were detected in extraintestinal tissues of patients with APECED, especially in medullary thymic epithelial cells. Some patients with APECED lacked Paneth cells and were seropositive for defensin-specific autoantibodies; the presence of autoantibodies correlated with frequent diarrhea. Aire(-/-) mice developed defensin-specific T cells. Adoptive transfer of these T cells to athymic mice resulted in T-cell infiltration of the gut, loss of Paneth cells, microbial dysbiosis, and the induction of T-helper 17 cell-mediated autoimmune responses resembling those observed in patients with APECED. CONCLUSIONS: In patients with APECED, loss of AIRE appears to cause an autoimmune response against enteric defensins and loss of Paneth cells. Aire(-/-) mice developed defensin-specific T cells that cause intestinal defects similar to those observed in patients with APECED. These findings provide a mechanism by which loss of AIRE-mediated immune tolerance leads to intestinal disorders in patients with APECED.


Subject(s)
Autoimmunity , Intestines/immunology , Polyendocrinopathies, Autoimmune/immunology , Transcription Factors/genetics , alpha-Defensins/immunology , Adolescent , Adult , Aged , Animals , Child , Child, Preschool , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Polyendocrinopathies, Autoimmune/complications , T-Lymphocytes/immunology , Young Adult , AIRE Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...