Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Nat Immunol ; 23(9): 1330-1341, 2022 09.
Article in English | MEDLINE | ID: mdl-35999392

ABSTRACT

Fibroblasts, the most abundant structural cells, exert homeostatic functions but also drive disease pathogenesis. Single-cell technologies have illuminated the shared characteristics of pathogenic fibroblasts in multiple diseases including autoimmune arthritis, cancer and inflammatory colitis. However, the molecular mechanisms underlying the disease-associated fibroblast phenotypes remain largely unclear. Here, we identify ETS1 as the key transcription factor governing the pathological tissue-remodeling programs in fibroblasts. In arthritis, ETS1 drives polarization toward tissue-destructive fibroblasts by orchestrating hitherto undescribed regulatory elements of the osteoclast differentiation factor receptor activator of nuclear factor-κB ligand (RANKL) as well as matrix metalloproteinases. Fibroblast-specific ETS1 deletion resulted in ameliorated bone and cartilage damage under arthritic conditions without affecting the inflammation level. Cross-tissue fibroblast single-cell data analyses and genetic loss-of-function experiments lent support to the notion that ETS1 defines the perturbation-specific fibroblasts shared among various disease settings. These findings provide a mechanistic basis for pathogenic fibroblast polarization and have important therapeutic implications.


Subject(s)
Arthritis, Rheumatoid , Fibroblasts , Proto-Oncogene Protein c-ets-1 , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Matrix Metalloproteinases/metabolism , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , RANK Ligand/genetics , Transcription Factors/metabolism
2.
Nat Immunol ; 21(10): 1172-1180, 2020 10.
Article in English | MEDLINE | ID: mdl-32839611

ABSTRACT

Fibroblasts are one of the most common but also neglected types of stromal cells, the heterogeneity of which underlies the specific function of tissue microenvironments in development and regeneration. In the thymus, autoreactive T cells are thought to be negatively selected by reference to the self-antigens expressed in medullary epithelial cells, but the contribution of other stromal cells to tolerance induction has been poorly examined. In the present study, we report a PDGFR+ gp38+ DPP4- thymic fibroblast subset that is required for T cell tolerance induction. The deletion of the lymphotoxin ß-receptor in thymic fibroblasts caused an autoimmune phenotype with decreased expression of tissue-restricted and fibroblast-specific antigens, offering insight into the long-sought target of lymphotoxin signaling in the context of the regulation of autoimmunity. Thus, thymic medullary fibroblasts play an essential role in the establishment of central tolerance by producing a diverse array of self-antigens.


Subject(s)
Fibroblasts/immunology , T-Lymphocytes/immunology , Thymus Gland/metabolism , Animals , Autoantigens/immunology , Autoimmunity , Cells, Cultured , Cellular Microenvironment , Clonal Selection, Antigen-Mediated , Dipeptidyl Peptidase 4/metabolism , Immune Tolerance , Lymphotoxin beta Receptor/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Thymus Gland/cytology
3.
Nat Immunol ; 19(11): 1265-1276, 2018 11.
Article in English | MEDLINE | ID: mdl-30323341

ABSTRACT

The methylation of arginine residues in proteins is a post-translational modification that contributes to a wide range of biological processes. Many cytokines involved in T cell development and activation utilize the common cytokine receptor γ-chain (γc) and the kinase JAK3 for signal transduction, but the regulatory mechanism that underlies the expression of these factors remains unclear. Here we found that the arginine methyltransferase PRMT5 was essential for the maintenance of invariant natural killer T cells (iNKT cells), CD4+ T cells and CD8+ T cells. T cell-specific deletion of Prmt5 led to a marked reduction in signaling via γc-family cytokines and a substantial loss of thymic iNKT cells, as well as a decreased number of peripheral CD4+ T cells and CD8+ T cells. PRMT5 induced the symmetric dimethylation of Sm proteins that promoted the splicing of pre-mRNA encoding γc and JAK3, and this critically contributed to the expression of γc and JAK3. Thus, arginine methylation regulates strength of signaling via γc-family cytokines by facilitating the expression of signal-transducing components.


Subject(s)
Arginine/metabolism , Interleukin Receptor Common gamma Subunit/immunology , Protein-Arginine N-Methyltransferases/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Interleukin Receptor Common gamma Subunit/metabolism , Methylation , Mice , Protein-Arginine N-Methyltransferases/immunology , T-Lymphocytes/metabolism
4.
Cell ; 163(4): 975-87, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26544942

ABSTRACT

Self-tolerance to immune reactions is established via promiscuous expression of tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs), leading to the elimination of T cells that respond to self-antigens. The transcriptional regulator Aire has been thought to be sufficient for the induction of TRAs, despite some indications that other factors may promote TRA expression in the thymus. Here, we show that the transcription factor Fezf2 directly regulates various TRA genes in mTECs independently of Aire. Mice lacking Fezf2 in mTECs displayed severe autoimmune symptoms, including the production of autoantibodies and inflammatory cell infiltration targeted to peripheral organs. These responses differed from those detected in Aire-deficient mice. Furthermore, Fezf2 expression and Aire expression are regulated by distinct signaling pathways and promote the expression of different classes of proteins. Thus, two independent factors, Fezf2 and Aire, permit the expression of TRAs in the thymus to ensure immune tolerance.


Subject(s)
DNA-Binding Proteins/metabolism , Immune Tolerance , Nerve Tissue Proteins/metabolism , Thymus Gland/immunology , Animals , Autoantigens/immunology , Autoimmunity , DNA-Binding Proteins/genetics , Humans , Mice , Nerve Tissue Proteins/genetics , Signal Transduction , Thymocytes/immunology , Transcription Factors/metabolism , AIRE Protein
5.
Nat Immunol ; 18(6): 675-682, 2017 06.
Article in English | MEDLINE | ID: mdl-28436956

ABSTRACT

Immunoglobulin A (IgA) maintains a symbiotic equilibrium with intestinal microbes. IgA induction in the gut-associated lymphoid tissues (GALTs) is dependent on microbial sampling and cellular interaction in the subepithelial dome (SED). However it is unclear how IgA induction is predominantly initiated in the SED. Here we show that previously unrecognized mesenchymal cells in the SED of GALTs regulate bacteria-specific IgA production and diversify the gut microbiota. Mesenchymal cells expressing the cytokine RANKL directly interact with the gut epithelium to control CCL20 expression and microfold (M) cell differentiation. The deletion of mesenchymal RANKL impairs M cell-dependent antigen sampling and B cell-dendritic cell interaction in the SED, which results in a reduction in IgA production and a decrease in microbial diversity. Thus, the subepithelial mesenchymal cells that serve as M cell inducers have a fundamental role in the maintenance of intestinal immune homeostasis.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Lymphoid Tissue/immunology , Mesenchymal Stem Cells/immunology , RANK Ligand/immunology , Animals , B-Lymphocytes/immunology , Biodiversity , Cell Differentiation/immunology , Chemokine CCL20/immunology , Dendritic Cells/immunology , Flow Cytometry , Gastrointestinal Microbiome/genetics , Germinal Center , Lymphoid Tissue/cytology , Mesenchymal Stem Cells/ultrastructure , Mice , Microscopy, Electron , RANK Ligand/genetics , RNA, Ribosomal, 16S/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
Int Immunol ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916145

ABSTRACT

The thymus is an organ required for T cell development and is also an eosinophil-rich organ; however, the nature and function of thymic eosinophils remain unclear. Here, we characterized the gene expression and differentiation mechanism of thymic eosinophils in mice. Thymic eosinophils showed a distinct gene expression profile compared with other organ-resident eosinophils. The number of thymic eosinophils was controlled by medullary thymic epithelial cells. In Rag-deficient mice, the unique gene expression signature of thymic eosinophils was lost but restored by pre-T cell receptor signaling, which induces CD4+ CD8+ thymocyte differentiation, indicating that T cell differentiation beyond the CD4- CD8- stage is necessary and sufficient for the induction of thymic eosinophils. These results demonstrate that thymic eosinophils are quantitatively and qualitatively regulated by medullary thymic epithelial cells and developing thymocytes, respectively, suggesting that thymic eosinophils are a distinct, thymus-specific cell subset, induced by interactions with thymic cells.

7.
Immunol Rev ; 302(1): 68-85, 2021 07.
Article in English | MEDLINE | ID: mdl-34096078

ABSTRACT

Fibroblasts have recently attracted attention as a key stromal component that controls the immune responses in lymphoid tissues. The thymus has a unique microenvironment comprised of a variety of stromal cells, including fibroblasts and thymic epithelial cells (TECs), the latter of which is known to be important for T cell development because of their ability to express self-antigens. Thymic fibroblasts contribute to thymus organogenesis during embryogenesis and form the capsule and medullary reticular network in the adult thymus. However, the immunological significance of thymic fibroblasts has thus far only been poorly elucidated. In this review, we will summarize the current views on the development and functions of thymic fibroblasts as revealed by new technologies such as multicolor flow cytometry and single cell-based transcriptome profiling. Furthermore, the recently discovered role of medullary fibroblasts in the establishment of T cell tolerance by producing a unique set of self-antigens will be highlighted.


Subject(s)
Fibroblasts , T-Lymphocytes , Cell Differentiation , Epithelial Cells , Lymphocyte Activation , Stromal Cells , Thymus Gland
8.
Physiol Rev ; 97(4): 1295-1349, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28814613

ABSTRACT

The immune and skeletal systems share a variety of molecules, including cytokines, chemokines, hormones, receptors, and transcription factors. Bone cells interact with immune cells under physiological and pathological conditions. Osteoimmunology was created as a new interdisciplinary field in large part to highlight the shared molecules and reciprocal interactions between the two systems in both heath and disease. Receptor activator of NF-κB ligand (RANKL) plays an essential role not only in the development of immune organs and bones, but also in autoimmune diseases affecting bone, thus effectively comprising the molecule that links the two systems. Here we review the function, gene regulation, and signal transduction of osteoimmune molecules, including RANKL, in the context of osteoclastogenesis as well as multiple other regulatory functions. Osteoimmunology has become indispensable for understanding the pathogenesis of a number of diseases such as rheumatoid arthritis (RA). We review the various osteoimmune pathologies, including the bone destruction in RA, in which pathogenic helper T cell subsets [such as IL-17-expressing helper T (Th17) cells] induce bone erosion through aberrant RANKL expression. We also focus on cellular interactions and the identification of the communication factors in the bone marrow, discussing the contribution of bone cells to the maintenance and regulation of hematopoietic stem and progenitors cells. Thus the time has come for a basic reappraisal of the framework for understanding both the immune and bone systems. The concept of a unified osteoimmune system will be absolutely indispensable for basic and translational approaches to diseases related to bone and/or the immune system.


Subject(s)
Immunity , Skeleton/immunology , Allergy and Immunology , Animals , Arthritis, Rheumatoid/immunology , Cell Communication , Hematopoietic Stem Cells/physiology , Humans , Osteoclasts/metabolism , Osteology , Osteoprotegerin/metabolism , RANK Ligand/immunology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction , Skeleton/metabolism
9.
Ann Rheum Dis ; 82(9): 1153-1161, 2023 09.
Article in English | MEDLINE | ID: mdl-37400117

ABSTRACT

OBJECTIVES: Recent studies demonstrate that extracellular-released aminoacyl-tRNA synthetases (aaRSs) play unique roles in immune responses and diseases. This study aimed to understand the role of extracellular aaRSs in the pathogenesis of rheumatoid arthritis (RA). METHODS: Primary macrophages and fibroblast-like synoviocytes were cultured with aaRSs. aaRS-induced cytokine production including IL-6 and TNF-α was detected by ELISA. Transcriptomic features of aaRS-stimulated macrophages were examined using RNA-sequencing. Serum and synovial fluid (SF) aaRS levels in patients with RA were assessed using ELISA. Peptidyl arginine deiminase (PAD) 4 release from macrophages stimulated with aaRSs was detected by ELISA. Citrullination of aaRSs by themselves was examined by immunoprecipitation and western blotting. Furthermore, aaRS inhibitory peptides were used for inhibition of arthritis in two mouse RA models, collagen-induced arthritis and collagen antibody-induced arthritis. RESULTS: All 20 aaRSs functioned as alarmin; they induced pro-inflammatory cytokines through the CD14-MD2-TLR4 axis. Stimulation of macrophages with aaRSs displayed persistent innate inflammatory responses. Serum and SF levels of many aaRSs increased in patients with RA compared with control subjects. Furthermore, aaRSs released PAD4 from living macrophages, leading to their citrullination. We demonstrate that aaRS inhibitory peptides suppress cytokine production and PAD4 release by aaRSs and alleviate arthritic symptoms in a mouse RA model. CONCLUSIONS: Our findings uncovered the significant role of aaRSs as a novel alarmin in RA pathogenesis, indicating that their blocking agents are potent antirheumatic drugs.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Animals , Mice , Alarmins , Cells, Cultured , Cytokines , Disease Models, Animal , Fibroblasts/pathology , Inflammation , Synovial Fluid , Humans
10.
Int Immunol ; 34(1): 45-52, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34687536

ABSTRACT

Medullary thymic epithelial cells (mTECs) help shape the thymic microenvironment for T-cell development by expressing a variety of peripheral tissue-restricted antigens (TRAs). The self-tolerance of T cells is established by negative selection of autoreactive T cells that bind to TRAs. To increase the diversity of TRAs, a fraction of mTECs terminally differentiates into distinct subsets resembling atypical types of epithelial cells in specific peripheral tissues. As such, thymic tuft cells that express peripheral tuft cell genes have recently emerged. Here, we show that the transcription factor SRY-box transcription factor 4 (Sox4) is highly expressed in mTECs and is essential for the development of thymic tuft cells. Mice lacking Sox4 specifically in TECs had a significantly reduced number of thymic tuft cells with no effect on the differentiation of other mTEC subsets, including autoimmune regulator (Aire)+ and Ccl21a+ mTECs. Furthermore, Sox4 expression was diminished in mice deficient in TEC-specific lymphotoxin ß receptor (LTßR), indicating a role for the LTßR-Sox4 axis in the differentiation of thymic tuft cells. Given that Sox4 promotes differentiation of peripheral tuft cells, our findings suggest that mTECs employ the same transcriptional program as peripheral epithelial cells. This mechanism may explain how mTECs diversify peripheral antigen expression to project an immunological self within the thymic medulla.


Subject(s)
Lymphotoxin beta Receptor/genetics , SOXC Transcription Factors/genetics , Thymus Gland/immunology , Animals , Cell Differentiation/immunology , Lymphotoxin beta Receptor/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , SOXC Transcription Factors/immunology , Signal Transduction/genetics , Thymus Gland/cytology
11.
Immunity ; 36(3): 427-37, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22425250

ABSTRACT

The thymic medulla provides a specialized microenvironment for the negative selection of T cells, with the presence of autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) during the embryonic-neonatal period being both necessary and sufficient to establish long-lasting tolerance. Here we showed that emergence of the first cohorts of Aire(+) mTECs at this key developmental stage, prior to αß T cell repertoire selection, was jointly directed by Rankl(+) lymphoid tissue inducer cells and invariant Vγ5(+) dendritic epidermal T cell (DETC) progenitors that are the first thymocytes to express the products of gene rearrangement. In turn, generation of Aire(+) mTECs then fostered Skint-1-dependent, but Aire-independent, DETC progenitor maturation and the emergence of an invariant DETC repertoire. Hence, our data attributed a functional importance to the temporal development of Vγ5(+) γδ T cells during thymus medulla formation for αß T cell tolerance induction and demonstrated a Rank-mediated reciprocal link between DETC and Aire(+) mTEC maturation.


Subject(s)
Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Transcription Factors/immunology , Animals , Cell Differentiation/immunology , Cellular Microenvironment , Epithelial Cells/immunology , Female , Fetus/cytology , Fetus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Signal Transduction/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Transcription Factors/deficiency , Transcription Factors/genetics , AIRE Protein
12.
J Allergy Clin Immunol ; 143(5): 1878-1891, 2019 05.
Article in English | MEDLINE | ID: mdl-30339851

ABSTRACT

BACKGROUND: Ras homolog gene family H (RhoH) is a membrane-bound adaptor protein involved in proximal T-cell receptor signaling. Therefore RhoH plays critical roles in the differentiation of T cells; however, the function of RhoH in the effecter phase of the T-cell response has not been fully characterized. OBJECTIVE: We sought to explore the role of RhoH in inflammatory immune responses and investigated the involvement of RhoH in the pathogenesis of psoriasis. METHODS: We analyzed effector T-cell and systemic inflammation in wild-type and RhoH-null mice. RhoH expression in T cells in human PBMCs was quantified by using RT-PCR. RESULTS: RhoH deficiency in mice induced TH17 polarization during effector T-cell differentiation, thereby inducing psoriasis-like chronic dermatitis. Ubiquitin protein ligase E3 component N-recognin 5 (Ubr5) and nuclear receptor subfamily 2 group F member 6 (Nr2f6) expression levels decreased in RhoH-deficient T cells, resulting in increased protein levels and DNA binding activity of retinoic acid-related orphan receptor γt. The consequential increase in IL-17 and IL-22 production induced T cells to differentiate into TH17 cells. Furthermore, IL-22 binding protein/Fc chimeric protein reduced psoriatic inflammation in RhoH-deficient mice. Expression of RhoH in T cells was lower in patients with psoriasis with very severe symptoms. CONCLUSION: Our results indicate that RhoH inhibits TH17 differentiation and thereby plays a role in the pathogenesis of psoriasis. Additionally, IL-22 binding protein has therapeutic potential for the treatment of psoriasis.


Subject(s)
Dermatitis/metabolism , Interleukins/metabolism , Psoriasis/metabolism , Th17 Cells/immunology , Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chronic Disease , Dermatitis/drug therapy , Dermatitis/genetics , Disease Models, Animal , Humans , Interleukins/genetics , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Psoriasis/drug therapy , Psoriasis/genetics , Receptors, Interleukin/therapeutic use , Repressor Proteins/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , rho GTP-Binding Proteins/genetics , Interleukin-22
13.
Int Immunol ; 30(7): 301-309, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29718261

ABSTRACT

γδT cells develop in the thymus and play important roles in protection against infection and tumor development, but the mechanisms by which the thymic microenvironment supports γδT cell differentiation remain largely unclear. Skint1, a B7-related protein expressed in thymic epithelial cells, was shown to be essential for the development of mouse Vγ5Vδ1 γδT cells. The Skint family in mouse consists of 11 members, Skint1-11. Here we generated mutant mice lacking the entire genomic region that contains all of the Skint genes. These mice exhibited a marked reduction of Vγ5Vδ1 γδT cells in the thymus and skin, but surprisingly, had normal development of other γδT cell subsets and leukocytes including αßT, B and myeloid cells. This phenotype is essentially identical to that of Skint1-deficient mice. These results indicate that the Skint family exerts an exclusive function in regulating the development of Vγ5Vδ1 γδT cells and is dispensable for development of other leukocytes.


Subject(s)
Immunoglobulins/deficiency , Immunoglobulins/genetics , Animals , Immunoglobulins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
14.
Immunity ; 32(1): 29-40, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20045355

ABSTRACT

How self-peptides displayed in the thymus contribute to the development of immunocompetent and self-protective T cells is largely unknown. In contrast, the role of thymic self-peptides in eliminating self-reactive T cells and thereby preventing autoimmunity is well established. A type of proteasome, termed thymoproteasome, is specifically expressed by thymic cortical epithelial cells (cTECs) and is required for the generation of optimal cellularity of CD8+ T cells. Here, we show that cTECs displayed thymoproteasome-specific peptide-MHC class I complexes essential for the positive selection of major and diverse repertoire of MHC class I-restricted T cells. CD8+ T cells generated in the absence of thymoproteasomes displayed a markedly altered T cell receptor repertoire that was defective in both allogeneic and antiviral responses. These results demonstrate that thymoproteasome-dependent self-peptide production is required for the development of an immunocompetent repertoire of CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Epithelial Cells/immunology , Proteasome Endopeptidase Complex/immunology , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Epithelial Cells/metabolism , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Self Tolerance/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
15.
Biochem Biophys Res Commun ; 496(1): 25-30, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29291408

ABSTRACT

Fine regulation of the Ras/mitogen-activating protein kinase (MAPK) pathway is crucial in controlling the survival, proliferation, and development of various types of cells. Ras-activating protein-like 3 (Rasal3) is a T cell-specific Ras GTPase-activating protein that negatively regulates T cell receptor (TCR)-induced activation of Ras/MAPK pathway. Rasal3-deficient mice showed a decreased number of naive T cells because Rasal3 is required for the survival of naive T cells. In the current study, we observed ameliorated Type1 T helper (Th1) cell- and Type2 T helper (Th2) cell-dependent contact hypersensitivity reactions in Rasal3-deficient mice, along with a marked shortage of T cells at regional lymph node. Activated Rasal3-deficient T cells showed an increased cell death with reduced Bcl2 expression, suggesting that Rasal3 is required for the survival of not only naïve T cells but also activated T cells. Collectively, Rasal3 controls the magnitude of inflammatory responses through the survival of both naive T cells and activated T cells in vivo.


Subject(s)
Cell Survival/immunology , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , ras GTPase-Activating Proteins/immunology , Animals , Mice , Mice, Inbred C57BL
16.
Biochem Biophys Res Commun ; 493(1): 437-443, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28882590

ABSTRACT

The gut-associated lymphoid tissues (GALTs), including Peyer's patches (PPs), cryptopatches (CPs) and isolated lymphoid follicles (ILFs), establish a host-microbe symbiosis by the promotion of immune reactions against gut microbes. Microfold cell inducer (MCi) cells in GALTs are the recently identified mesenchymal cells that express the cytokine RANKL and initiate bacteria-specific immunoglobulin A (IgA) production via induction of microfold (M) cell differentiation. In the previous study, the Twist2-Cre driver was utilized for gene deletion in mesenchymal cells including MCi cells. In order to investigate MCi cells more extensively, it will be necessary to develop experimental tools in addition to the Twist2-Cre driver mice and characterize such drivers in specificity and efficiency. Here we show that M cell differentiation and IgA production are impaired in the targeted deletion of RANKL by the Col6a1-Cre driver. We compared Col6a1-Cre with Twist2-Cre in terms of the specificity for mesenchymal cells in GALTs. Col6a1-Cre CAG-CAT-EGFP mice exhibited EGFP expression in podoplanin+CD31- cells including MCi cells, while Twist2-Cre mice were shown to target endothelial cells and podoplanin+CD31- cells. Tnfsf11fl/ΔCol6a1-Cre mice exhibited the absence of M cells and severe IgA reduction together with an alteration in gut microbial composition. Moreover, we analyzed germ free mice to test whether changes in the microbiota are the cause of M cell deficiency. M cell differentiation was normal in the CPs/ILFs of germ free mice, indicating that MCi cells induce M cells independently of microbial colonization. This study demonstrates that Col6a1-Cre driver mice are as useful as Twist2-Cre driver mice for functional analyses of GALT-resident mesenchymal cells, including MCi cells.


Subject(s)
Collagen Type VI/genetics , Integrases/genetics , Intestinal Mucosa/immunology , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/immunology , T-Lymphocytes, Helper-Inducer/physiology , Animals , Cells, Cultured , Gene Deletion , Mice , Mice, Inbred C57BL
17.
Immunity ; 29(3): 451-63, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18799151

ABSTRACT

Medullary thymic epithelial cells (mTECs) are specialized for inducing central immunological tolerance to self-antigens. To accomplish this, mTECs must adopt a mature phenotype characterized by expression of the autoimmune regulator Aire, which activates the transcription of numerous genes encoding tissue-restricted self-antigens. The mechanisms that control mature Aire(+) mTEC development in the postnatal thymus remain poorly understood. We demonstrate here that, although either CD4(+) or CD8(+) thymocytes are sufficient to sustain formation of a well-defined medulla, expansion of the mature mTEC population requires autoantigen-specific interactions between positively selected CD4(+) thymocytes bearing autoreactive T cell receptor (TCR) and mTECs displaying cognate self-peptide-MHC class II complexes. These interactions also involve the engagement of CD40 on mTECs by CD40L induced on the positively selected CD4(+) thymocytes. This antigen-specific TCR-MHC class II-mediated crosstalk between CD4(+) thymocytes and mTECs defines a unique checkpoint in thymic stromal development that is pivotal for generating a mature mTEC population competent for ensuring central T cell tolerance.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Mice , Mice, Knockout , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Self Tolerance , Thymus Gland/cytology , Thymus Gland/metabolism , Trans-Activators/immunology , Trans-Activators/metabolism , Transcription Factors/immunology , AIRE Protein
18.
Immunity ; 29(3): 423-37, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18799149

ABSTRACT

Medullary thymic epithelial cells (mTECs) establish T cell self-tolerance through the expression of autoimmune regulator (Aire) and peripheral tissue-specific self-antigens. However, signals underlying mTEC development remain largely unclear. Here, we demonstrate crucial regulation of mTEC development by receptor activator of NF-kappaB (RANK) and CD40 signals. Whereas only RANK signaling was essential for mTEC development during embryogenesis, in postnatal mice, cooperation between CD40 and RANK signals was required for mTEC development to successfully establish the medullary microenvironment. Ligation of RANK or CD40 on fetal thymic stroma in vitro induced mTEC development in a tumor necrosis factor-associated factor 6 (TRAF6)-, NF-kappaB inducing kinase (NIK)-, and IkappaB kinase beta (IKKbeta)-dependent manner. These results show that developmental-stage-dependent cooperation between RANK and CD40 promotes mTEC development, thereby establishing self-tolerance.


Subject(s)
CD40 Antigens/metabolism , CD40 Ligand/metabolism , Epithelial Cells/cytology , Epithelial Cells/immunology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Self Tolerance , Thymus Gland/cytology , Animals , Autoimmunity , CD40 Antigens/deficiency , Cell Differentiation , Epithelial Cells/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , RANK Ligand/deficiency , RANK Ligand/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Thymus Gland/embryology , Thymus Gland/immunology , Thymus Gland/physiology , NF-kappaB-Inducing Kinase
19.
Immunity ; 29(3): 438-50, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18799150

ABSTRACT

The thymic medulla provides a microenvironment where medullary thymic epithelial cells (mTECs) express autoimmune regulator and diverse tissue-restricted genes, contributing to launching self-tolerance. Positive selection is essential for thymic medulla formation via a previously unknown mechanism. Here we show that the cytokine RANK ligand (RANKL) was produced by positively selected thymocytes and regulated the cellularity of mTEC by interacting with RANK and osteoprotegerin. Forced expression of RANKL restored thymic medulla in mice lacking positive selection, whereas RANKL perturbation impaired medulla formation. These results indicate that RANKL produced by positively selected thymocytes is responsible for fostering thymic medulla formation, thereby establishing central tolerance.


Subject(s)
Epithelial Cells/immunology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Autoimmunity , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mice , Osteoprotegerin/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Self Tolerance , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , AIRE Protein
20.
EMBO Rep ; 16(5): 638-53, 2015 May.
Article in English | MEDLINE | ID: mdl-25770130

ABSTRACT

The thymus provides a specialized microenvironment in which distinct subsets of thymic epithelial cells (TECs) support T-cell development. Here, we describe the significance of cortical TECs (cTECs) in T-cell development, using a newly established mouse model of cTEC deficiency. The deficiency of mature cTECs caused a massive loss of thymic cellularity and impaired the development of αßT cells and invariant natural killer T cells. Unexpectedly, the differentiation of certain γδT-cell subpopulations-interleukin-17-producing Vγ4 and Vγ6 cells-was strongly dysregulated, resulting in the perturbation of γδT-mediated inflammatory responses in peripheral tissues. These findings show that cTECs contribute to the shaping of the TCR repertoire, not only of "conventional" αßT cells but also of inflammatory "innate" γδT cells.


Subject(s)
Epithelium/metabolism , Interleukin-17/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism , Animals , Cell Differentiation , Cell Survival/genetics , DNA Mutational Analysis , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/immunology , Female , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/immunology , Thymus Gland/pathology
SELECTION OF CITATIONS
SEARCH DETAIL