1
|
Monteiro CJ, Duarte MJ, Machado MCV, Mascarenhas RS, Palma PVB, García HDM, Nakaya HI, Cunha TM, Donadi EA, Passos GA. The single-cell transcriptome of mTECs and CD4 + thymocytes under adhesion revealed heterogeneity of mTECs and a network controlled by Aire and lncRNAs. Front Immunol 2024; 15:1376655. [PMID: 39328409 PMCID: PMC11425717 DOI: 10.3389/fimmu.2024.1376655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/05/2024] [Indexed: 09/28/2024] Open
Abstract
To further understand the impact of deficiency of the autoimmune regulator (Aire) gene during the adhesion of medullary thymic epithelial cells (mTECs) to thymocytes, we sequenced single-cell libraries (scRNA-seq) obtained from Aire wild-type (WT) (Airewt/wt ) or Aire-deficient (Airewt/mut ) mTECs cocultured with WT single-positive (SP) CD4+ thymocytes. Although the libraries differed in their mRNA and long noncoding RNA (lncRNA) profiles, indicating that mTECs were heterogeneous in terms of their transcriptome, UMAP clustering revealed that both mTEC lines expressed their specific markers, i.e., Epcam, Itgb4, Itga6, and Casp3 in resting mTECs and Ccna2, Pbk, and Birc5 in proliferative mTECs. Both cocultured SP CD4+ thymocytes remained in a homogeneous cluster expressing the Il7r and Ccr7 markers. Comparisons of the two types of cocultures revealed the differential expression of mRNAs that encode transcription factors (Zfpm2, Satb1, and Lef1), cell adhesion genes (Itgb1) in mTECs, and Themis in thymocytes, which is associated with the regulation of positive and negative selection. At the single-cell sequencing resolution, we observed that Aire acts on both Aire WT and Aire-deficient mTECs as an upstream controller of mRNAs, which encode transcription factors or adhesion proteins that, in turn, are posttranscriptionally controlled by lncRNAs, for example, Neat1, Malat1, Pvt1, and Dancr among others. Under Aire deficiency, mTECs dysregulate the expression of MHC-II, CD80, and CD326 (EPCAM) protein markers as well as metabolism and cell cycle-related mRNAs, which delay the cell cycle progression. Moreover, when adhered to mTECs, WT SP CD4+ or CD8+ thymocytes modulate the expression of cell activation proteins, including CD28 and CD152/CTLA4, and the expression of cellular metabolism mRNAs. These findings indicate a complex mechanism through which an imbalance in Aire expression can affect mTECs and thymocytes during adhesion.
Collapse
Affiliation(s)
- Cíntia J. Monteiro
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Max J. Duarte
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Mayara Cristina V. Machado
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Romário S. Mascarenhas
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Patrícia V. Bonini Palma
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | | | - Helder I. Nakaya
- Research Institute, Albert Einstein Israeli Hospital, São Paulo, SP, Brazil
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thiago M. Cunha
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Eduardo A. Donadi
- Department of Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Geraldo A. Passos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
2
|
Weiler P, Lange M, Klein M, Pe'er D, Theis F. CellRank 2: unified fate mapping in multiview single-cell data. Nat Methods 2024; 21:1196-1205. [PMID: 38871986 PMCID: PMC11239496 DOI: 10.1038/s41592-024-02303-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 05/09/2024] [Indexed: 06/15/2024]
Abstract
Single-cell RNA sequencing allows us to model cellular state dynamics and fate decisions using expression similarity or RNA velocity to reconstruct state-change trajectories; however, trajectory inference does not incorporate valuable time point information or utilize additional modalities, whereas methods that address these different data views cannot be combined or do not scale. Here we present CellRank 2, a versatile and scalable framework to study cellular fate using multiview single-cell data of up to millions of cells in a unified fashion. CellRank 2 consistently recovers terminal states and fate probabilities across data modalities in human hematopoiesis and endodermal development. Our framework also allows combining transitions within and across experimental time points, a feature we use to recover genes promoting medullary thymic epithelial cell formation during pharyngeal endoderm development. Moreover, we enable estimating cell-specific transcription and degradation rates from metabolic-labeling data, which we apply to an intestinal organoid system to delineate differentiation trajectories and pinpoint regulatory strategies.
Collapse
Affiliation(s)
- Philipp Weiler
- Institute of Computational Biology, Department of Computational Health, Helmholtz Munich, Munich, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Marius Lange
- Institute of Computational Biology, Department of Computational Health, Helmholtz Munich, Munich, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Michal Klein
- Institute of Computational Biology, Department of Computational Health, Helmholtz Munich, Munich, Germany
- Machine Learning Research, Apple, Paris, France
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Fabian Theis
- Institute of Computational Biology, Department of Computational Health, Helmholtz Munich, Munich, Germany.
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany.
| |
Collapse
|
3
|
Aytekin ES, Cagdas D. APECED and the place of AIRE in the puzzle of the immune network associated with autoimmunity. Scand J Immunol 2023; 98:e13299. [PMID: 38441333 DOI: 10.1111/sji.13299] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 03/07/2024]
Abstract
In the last 20 years, discoveries about the autoimmune regulator (AIRE) protein and its critical role in immune tolerance have provided fundamental insights into understanding the molecular basis of autoimmunity. This review provides a comprehensive overview of the effect of AIRE on immunological tolerance and the characteristics of autoimmune diseases in Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy (APECED), which is caused by biallelic AIRE mutations. A better understanding of the immunological mechanisms of AIRE deficiency may enlighten immune tolerance mechanisms and new diagnostic and treatment strategies for autoimmune diseases. Considering that not all clinical features of APECED are present in a given follow-up period, the diagnosis is not easy in a patient at the first visit. Longer follow-up and a multidisciplinary approach are essential for diagnosis. It is challenging to prevent endocrine and other organ damage compared with other diseases associated with multiple autoimmunities, such as FOXP3, LRBA, and CTLA4 deficiencies. Unfortunately, no curative therapy like haematopoietic stem cell transplantation or specific immunomodulation is present that is successful in the treatment.
Collapse
Affiliation(s)
- Elif Soyak Aytekin
- Pediatric Allergy and Immunology, Department of Pediatrics, SBU Dr. Sami Ulus Children Hospital, Ankara, Turkey
| | - Deniz Cagdas
- Division of Pediatric Immunology, Department of Pediatrics, Ihsan Dogramaci Children`s Hospital, Institute of Child Health, Hacettepe University Medical School, Ankara, Turkey
| |
Collapse
|
4
|
Villegas JA, Gradolatto A, Truffault F, Roussin R, Berrih-Aknin S, Le Panse R, Dragin N. Cultured Human Thymic-Derived Cells Display Medullary Thymic Epithelial Cell Phenotype and Functionality. Front Immunol 2018; 9:1663. [PMID: 30083154 PMCID: PMC6064927 DOI: 10.3389/fimmu.2018.01663] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/04/2018] [Indexed: 12/13/2022] Open
Abstract
Thymic epithelial cells are one of the main components of the thymic microenvironment required for T-cell development. In this work, we describe an efficient method free of enzymatic and Facs-sorted methods to culture human medullary thymic epithelial cells without affecting the cell phenotypic, physiologic and functional features. Human medulla thymic epithelial cells (mTECs) are obtained by culturing thymic biopsies explants. After 7 days of primo-culture, mTECs keep their ability to express key molecules involved in immune tolerance processes such as autoimmune regulator, tissue-specific antigens, chemokines, and cytokines. In addition, the cells sensor their cultured environment and consequently adjust their gene expression network. Therefore, we describe and provide a human mTEC model that may be used to test the effect of various molecules on thymic epithelial cell homeostasis and physiology. This method should allow the investigations of the specificities and the knowledge of human mTECs in normal or pathological conditions and therefore discontinue the extrapolations done on the murine models.
Collapse
Affiliation(s)
- José A Villegas
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | - Angeline Gradolatto
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | - Frédérique Truffault
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | | | - Sonia Berrih-Aknin
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | - Rozen Le Panse
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | - Nadine Dragin
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France.,Inovarion, Paris, France
| |
Collapse
|
5
|
Abstract
About two decades ago, cloning of the autoimmune regulator (AIRE) gene materialized one of the most important actors on the scene of self-tolerance. Thymic transcription of genes encoding tissue-specific antigens (ts-ags) is activated by AIRE protein and embodies the essence of thymic self-representation. Pathogenic AIRE variants cause the autoimmune polyglandular syndrome type 1, which is a rare and complex disease that is gaining attention in research on autoimmunity. The animal models of disease, although not identically reproducing the human picture, supply fundamental information on mechanisms and extent of AIRE action: thanks to its multidomain structure, AIRE localizes to chromatin enclosing the target genes, binds to histones, and offers an anchorage to multimolecular complexes involved in initiation and post-initiation events of gene transcription. In addition, AIRE enhances mRNA diversity by favoring alternative mRNA splicing. Once synthesized, ts-ags are presented to, and cause deletion of the self-reactive thymocyte clones. However, AIRE function is not restricted to the activation of gene transcription. AIRE would control presentation and transfer of self-antigens for thymic cellular interplay: such mechanism is aimed at increasing the likelihood of engagement of the thymocytes that carry the corresponding T-cell receptors. Another fundamental role of AIRE in promoting self-tolerance is related to the development of thymocyte anergy, as thymic self-representation shapes at the same time the repertoire of regulatory T cells. Finally, AIRE seems to replicate its action in the secondary lymphoid organs, albeit the cell lineage detaining such property has not been fully characterized. Delineation of AIRE functions adds interesting data to the knowledge of the mechanisms of self-tolerance and introduces exciting perspectives of therapeutic interventions against the related diseases.
Collapse
Affiliation(s)
- Roberto Perniola
- Department of Pediatrics, Neonatal Intensive Care, Vito Fazzi Regional Hospital, Lecce, Italy
| |
Collapse
|
6
|
Song Y, Sullivan T, Klarmann K, Gilbert D, O’Sullivan TN, Lu L, Wang S, Haines DC, Van Dyke T, Keller JR. RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice. PLoS One 2017; 12:e0171510. [PMID: 28158249 PMCID: PMC5291521 DOI: 10.1371/journal.pone.0171510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/20/2017] [Indexed: 11/25/2022] Open
Abstract
Thymic epithelial cells (TEC), as part of thymic stroma, provide essential growth factors/cytokines and self-antigens to support T cell development and selection. Deletion of Rb family proteins in adult thymic stroma leads to T cell hyperplasia in vivo. To determine whether deletion of Rb specifically in keratin (K) 18 positive TEC was sufficient for thymocyte hyperplasia, we conditionally inactivated Rb and its family members p107 and p130 in K18+ TEC in genetically engineered mice (TgK18GT121; K18 mice). We found that thymocyte hyperproliferation was induced in mice with Rb inactivation in K18+ TEC, while normal T cell development was maintained; suggesting that inactivation of Rb specifically in K18+ TEC was sufficient and responsible for the phenotype. Transplantation of wild type bone marrow cells into mice with Rb inactivation in K18+ TEC resulted in donor T lymphocyte hyperplasia confirming the non-cell autonomous requirement for Rb proteins in K18+ TEC in regulating T cell proliferation. Our data suggests that thymic epithelial cells play an important role in regulating lymphoid proliferation and thymus size.
Collapse
Affiliation(s)
- Yurong Song
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Teresa Sullivan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Kimberly Klarmann
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Debra Gilbert
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - T. Norene O’Sullivan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Lucy Lu
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Sophie Wang
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Diana C. Haines
- Pathology/ Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Terry Van Dyke
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Jonathan R. Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
7
|
β-catenin activation drives thymoma initiation and progression in mice. Oncotarget 2016; 6:13978-93. [PMID: 26101855 PMCID: PMC4546445 DOI: 10.18632/oncotarget.4368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/01/2015] [Indexed: 11/25/2022] Open
Abstract
Thymoma is the most commonly identified cancer in the anterior mediastinum. To date, the causal mechanism that drives thymoma progression is not clear. Here, we generated K5-ΔN64Ctnnb1/ERT2 transgenic mice, which express an N-terminal deletion mutant of β-catenin fused to a mutated ligand-binding domain of estrogen receptor (ERT2) under the control of the bovine cytokeratin 5 (K5) promoter. The transgenic mouse lines named Tg1 and Tg4 were characterized. Forced expression of ΔN64Ctnnb1/ERT2 in the Tg1 and Tg4 mice developed small thymoma lesions in response to tamoxifen treatment. In the absence of tamoxifen, the Tg1 mice exhibited leaky activation of β-catenin, which activated the TOP-Gal transgene and Wnt/β-catenin-targeted genes. As the Tg1 mice aged in the absence of tamoxifen, manifest thymomas were found at 10-12 months. Interestingly, we detected loss of AIRE and increase of p63 in the thymomas of Tg1 mice, similar to that observed in human thymomas. Moreover, the β5t protease subunit, which was reported as a differential marker for human type B3 thymoma, was expressed in the Tg1 thymomas. Thus, the Tg1 mice generated in this study accurately mimic the characteristics of human thymomas and may serve as a model for understanding thymoma pathogenesis.
Collapse
|
8
|
Dragin N, Bismuth J, Cizeron-Clairac G, Biferi MG, Berthault C, Serraf A, Nottin R, Klatzmann D, Cumano A, Barkats M, Le Panse R, Berrih-Aknin S. Estrogen-mediated downregulation of AIRE influences sexual dimorphism in autoimmune diseases. J Clin Invest 2016; 126:1525-37. [PMID: 26999605 PMCID: PMC4811157 DOI: 10.1172/jci81894] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 01/21/2016] [Indexed: 01/01/2023] Open
Abstract
Autoimmune diseases affect 5% to 8% of the population, and females are more susceptible to these diseases than males. Here, we analyzed human thymic transcriptome and revealed sex-associated differences in the expression of tissue-specific antigens that are controlled by the autoimmune regulator (AIRE), a key factor in central tolerance. We hypothesized that the level of AIRE is linked to sexual dimorphism susceptibility to autoimmune diseases. In human and mouse thymus, females expressed less AIRE (mRNA and protein) than males after puberty. These results were confirmed in purified murine thymic epithelial cells (TECs). We also demonstrated that AIRE expression is related to sexual hormones, as male castration decreased AIRE thymic expression and estrogen receptor α-deficient mice did not show a sex disparity for AIRE expression. Moreover, estrogen treatment resulted in downregulation of AIRE expression in cultured human TECs, human thymic tissue grafted to immunodeficient mice, and murine fetal thymus organ cultures. AIRE levels in human thymus grafted in immunodeficient mice depended upon the sex of the recipient. Estrogen also upregulated the number of methylated CpG sites in the AIRE promoter. Together, our results indicate that in females, estrogen induces epigenetic changes in the AIRE gene, leading to reduced AIRE expression under a threshold that increases female susceptibility to autoimmune diseases.
Collapse
Affiliation(s)
- Nadine Dragin
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Jacky Bismuth
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | | | - Maria Grazia Biferi
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Claire Berthault
- INSERM U668, Unit for Lymphopoiesis, Immunology Department, Pasteur Institute, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Alain Serraf
- Hôpital Marie Lannelongue, Le Plessis–Robinson, France
| | - Rémi Nottin
- Hôpital Marie Lannelongue, Le Plessis–Robinson, France
| | - David Klatzmann
- Assistance Publique – Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Biotherapy, Paris, France
| | - Ana Cumano
- INSERM U668, Unit for Lymphopoiesis, Immunology Department, Pasteur Institute, Paris, France
| | - Martine Barkats
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Rozen Le Panse
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| |
Collapse
|
9
|
Nitta T, Suzuki H. Thymic stromal cell subsets for T cell development. Cell Mol Life Sci 2016; 73:1021-37. [PMID: 26825337 PMCID: PMC11108406 DOI: 10.1007/s00018-015-2107-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/26/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022]
Abstract
The thymus provides a specialized microenvironment in which a variety of stromal cells of both hematopoietic and non-hematopoietic origin regulate development and repertoire selection of T cells. Recent studies have been unraveling the inter- and intracellular signals and transcriptional networks for spatiotemporal regulation of development of thymic stromal cells, mainly thymic epithelial cells (TECs), and the molecular mechanisms of how different TEC subsets control T cell development and selection. TECs are classified into two functionally different subsets: cortical TECs (cTECs) and medullary TECs (mTECs). cTECs induce positive selection of diverse and functionally distinct T cells by virtue of unique antigen-processing systems, while mTECs are essential for establishing T cell tolerance via ectopic expression of peripheral tissue-restricted antigens and cooperation with dendritic cells. In addition to reviewing the role of the thymic stroma in conventional T cell development, we will discuss recently discovered novel functions of TECs in the development of unconventional T cells, such as natural killer T cells and γδT cells.
Collapse
Affiliation(s)
- Takeshi Nitta
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, 272-8516, Japan.
| | - Harumi Suzuki
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, 272-8516, Japan.
| |
Collapse
|
10
|
Kawano H, Nishijima H, Morimoto J, Hirota F, Morita R, Mouri Y, Nishioka Y, Matsumoto M. Aire Expression Is Inherent to Most Medullary Thymic Epithelial Cells during Their Differentiation Program. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:5149-58. [PMID: 26503950 DOI: 10.4049/jimmunol.1501000] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/02/2015] [Indexed: 11/19/2022]
Abstract
Aire in medullary thymic epithelial cells (mTECs) plays an important role in the establishment of self-tolerance. Because Aire(+) mTECs appear to be a limited subset, they may constitute a unique lineage(s) among mTECs. An alternative possibility is that all mTECs are committed to express Aire in principle, but Aire expression by individual mTECs is conditional. To investigate this issue, we established a novel Aire reporter strain in which endogenous Aire is replaced by the human AIRE-GFP-Flag tag (Aire/hAGF-knockin) fusion gene. The hAGF reporter protein was produced and retained very efficiently within mTECs as authentic Aire nuclear dot protein. Remarkably, snapshot analysis revealed that mTECs expressing hAGF accounted for >95% of mature mTECs, suggesting that Aire expression does not represent a particular mTEC lineage(s). We confirmed this by generating Aire/diphtheria toxin receptor-knockin mice in which long-term ablation of Aire(+) mTECs by diphtheria toxin treatment resulted in the loss of most mature mTECs beyond the proportion of those apparently expressing Aire. These results suggest that Aire expression is inherent to all mTECs but may occur at particular stage(s) and/or cellular states during their differentiation, thus accounting for the broad impact of Aire on the promiscuous gene expression of mTECs.
Collapse
Affiliation(s)
- Hiroshi Kawano
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan; Department of Respiratory Medicine and Rheumatology, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima 770-8503, Japan; and
| | - Hitoshi Nishijima
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Junko Morimoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Fumiko Hirota
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan; Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology, Tokyo 100-0004, Japan
| | - Ryoko Morita
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan; Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology, Tokyo 100-0004, Japan
| | - Yasuhiro Mouri
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima 770-8503, Japan; and
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan; Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology, Tokyo 100-0004, Japan
| |
Collapse
|
11
|
Hu Z, Lancaster JN, Ehrlich LIR. The Contribution of Chemokines and Migration to the Induction of Central Tolerance in the Thymus. Front Immunol 2015; 6:398. [PMID: 26300884 PMCID: PMC4528182 DOI: 10.3389/fimmu.2015.00398] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/20/2015] [Indexed: 02/01/2023] Open
Abstract
As T cells develop, they migrate throughout the thymus where they undergo essential bi-directional signaling with stromal cells in distinct thymic microenvironments. Immature thymocyte progenitors are located in the thymic cortex. Following T cell receptor expression and positive selection, thymocytes undergo a dramatic transition: they become rapidly motile and relocate to the thymic medulla. Antigen-presenting cells (APCs) within the cortex and medulla display peptides derived from a wide array of self-proteins, which promote thymocyte self-tolerance. If a thymocyte is auto-reactive against such antigens, it undergoes either negative selection, via apoptosis, or differentiation into the regulatory T cell lineage. This induction of central tolerance is critical for prevention of autoimmunity. Chemokines and adhesion molecules play an essential role in tolerance induction, as they promote migration of developing thymocytes through the different thymic microenvironments and enhance interactions with APCs displaying self-antigens. Herein, we review the contribution of chemokines and other regulators of thymocyte localization and motility to T cell development, with a focus on their contribution to the induction of central tolerance.
Collapse
Affiliation(s)
- Zicheng Hu
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| | - Jessica Naomi Lancaster
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| | - Lauren I R Ehrlich
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| |
Collapse
|
12
|
Lopes N, Ferrier P, Irla M. [Induction of central tolerance by the factor Aire: molecular and epigenetic regulation]. Med Sci (Paris) 2015; 31:742-7. [PMID: 26340833 DOI: 10.1051/medsci/20153108012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
The establishment of thymic central tolerance is a critical process to prevent the development of autoimmune diseases. Medullary thymic epithelial cells (mTEC) are essential to this process through the expression of the transcription factor Aire, which controls the transcription of many genes encoding tissue-restricted antigens. Mutations in the Aire gene are responsible for a rare autoimmune disorder called APECED (autoimmune polyendocrinopathy candidiasis ectodermal dystrophy). This review summarizes our current knowledge on the mode of action of Aire at the molecular and epigenetic levels in controlling the expression of tissue-restricted antigens. We also discuss recently described additional roles of this transcription factor in the induction of central T-cell tolerance.
Collapse
Affiliation(s)
- Noëlla Lopes
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France
| | - Pierre Ferrier
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France
| | - Magali Irla
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France
| |
Collapse
|
13
|
St-Pierre C, Trofimov A, Brochu S, Lemieux S, Perreault C. Differential Features of AIRE-Induced and AIRE-Independent Promiscuous Gene Expression in Thymic Epithelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:498-506. [PMID: 26034170 DOI: 10.4049/jimmunol.1500558] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/04/2015] [Indexed: 12/16/2023]
Abstract
Establishment of self-tolerance in the thymus depends on promiscuous expression of tissue-restricted Ags (TRA) by thymic epithelial cells (TEC). This promiscuous gene expression (pGE) is regulated in part by the autoimmune regulator (AIRE). To evaluate the commonalities and discrepancies between AIRE-dependent and -independent pGE, we analyzed the transcriptome of the three main TEC subsets in wild-type and Aire knockout mice. We found that the impact of AIRE-dependent pGE is not limited to generation of TRA. AIRE decreases, via non-cell autonomous mechanisms, the expression of genes coding for positive regulators of cell proliferation, and it thereby reduces the number of cortical TEC. In mature medullary TEC, AIRE-driven pGE upregulates non-TRA coding genes that enhance cell-cell interactions (e.g., claudins, integrins, and selectins) and are probably of prime relevance to tolerance induction. We also found that AIRE-dependent and -independent TRA present several distinctive features. In particular, relative to AIRE-induced TRA, AIRE-independent TRA are more numerous and show greater splicing complexity. Furthermore, we report that AIRE-dependent versus -independent TRA project nonredundant representations of peripheral tissues in the thymus.
Collapse
Affiliation(s)
- Charles St-Pierre
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; and
| | - Assya Trofimov
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; and Department of Computer Science and Operations Research, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Sylvie Brochu
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; and
| | - Sébastien Lemieux
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Computer Science and Operations Research, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; and
| |
Collapse
|
14
|
Niemi HJ, Laakso S, Salminen JT, Arstila TP, Tuulasvaara A. A normal T cell receptor beta CDR3 length distribution in patients with APECED. Cell Immunol 2015; 295:99-104. [DOI: 10.1016/j.cellimm.2015.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 02/18/2015] [Accepted: 03/13/2015] [Indexed: 10/23/2022]
|
15
|
Franckaert D, Schlenner SM, Heirman N, Gill J, Skogberg G, Ekwall O, Put K, Linterman MA, Dooley J, Liston A. Premature thymic involution is independent of structural plasticity of the thymic stroma. Eur J Immunol 2015; 45:1535-47. [PMID: 25627671 PMCID: PMC4670717 DOI: 10.1002/eji.201445277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/22/2014] [Accepted: 01/23/2015] [Indexed: 12/14/2022]
Abstract
The thymus is the organ devoted to T-cell production. The thymus undergoes multiple rounds of atrophy and redevelopment before degenerating with age in a process known as involution. This process is poorly understood, despite the influence the phenomenon has on peripheral T-cell numbers. Here we have investigated the FVB/N mouse strain, which displays premature thymic involution. We find multiple architectural and cellular features that precede thymic involution, including disruption of the epithelial–endothelial relationship and a progressive loss of pro-T cells. The architectural features, reminiscent of the human thymus, are intrinsic to the nonhematopoietic compartment and are neither necessary nor sufficient for thymic involution. By contrast, the loss of pro-T cells is intrinsic to the hematopoietic compartment, and is sufficient to drive premature involution. These results identify pro-T-cell loss as the main driver of premature thymic involution, and highlight the plasticity of the thymic stroma, capable of maintaining function across diverse interstrain architectures.
Collapse
Affiliation(s)
- Dean Franckaert
- Autoimmune Genetics Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Susan M Schlenner
- Autoimmune Genetics Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Nathalie Heirman
- Autoimmune Genetics Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Jason Gill
- Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Gabriel Skogberg
- Department of Rheumatology and Inflammation Research, Göteborg University, Gothenburg, Sweden
| | - Olov Ekwall
- Department of Rheumatology and Inflammation Research, Göteborg University, Gothenburg, Sweden
| | - Karen Put
- Autoimmune Genetics Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | | | - James Dooley
- Autoimmune Genetics Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Adrian Liston
- Autoimmune Genetics Laboratory, VIB, Leuven, Belgium
| |
Collapse
|
16
|
Suzuki S, Suzuki M, Nakai M, Sembon S, Fuchimoto D, Onishi A. Transcriptional and histological analyses of the thymic developmental process in the fetal pig. Exp Anim 2014; 63:215-25. [PMID: 24770647 PMCID: PMC4160976 DOI: 10.1538/expanim.63.215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The humanized pig model, in which human cells or tissues can be functionally maintained
in pigs, can be an invaluable tool for human medical research. Although the recent
development of immunodeficient pigs has opened the door for the development of such a
model, the efficient engraftment and differentiation of human cells may be difficult to
achieve. The transplantation of human cells into fetal pigs, whose immune system is
immature, will ameliorate this problem. Therefore, we examined the development of porcine
fetal thymus, which is critical for the establishment of the immune system. We first
analyzed the levels of mRNA expression of genes that are relevant to the function of
thymic epithelial cells or thymocytes in whole thymi from 35 to 85 days of gestation (DG)
and at 2 days postpartum (DP) by quantitative RT-PCR. In addition, immunohistochemical
analyses of thymic epithelial cells from DG35 to DG55 and DP2 were performed. These
analyses showed that the thymic cortex was formed as early as DG35, and thymic medulla
gradually developed from DG45 to DG55. These findings suggested that, at least before
DG45, the thymus do not differentiate to form fully functional T cells.
Collapse
Affiliation(s)
- Shunichi Suzuki
- Transgenic Pig Research Unit, National Institute of Agrobiological Sciences, 2 Ikenodai, Tsukuba,Ibaraki 305-0901, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Sawanobori Y, Ueta H, Dijkstra CD, Park CG, Satou M, Kitazawa Y, Matsuno K. Three distinct subsets of thymic epithelial cells in rats and mice defined by novel antibodies. PLoS One 2014; 9:e109995. [PMID: 25334032 PMCID: PMC4204869 DOI: 10.1371/journal.pone.0109995] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 09/04/2014] [Indexed: 01/04/2023] Open
Abstract
AIM Thymic epithelial cells (TECs) are thought to play an essential role in T cell development and have been detected mainly in mice using lectin binding and antibodies to keratins. Our aim in the present study was to create a precise map of rat TECs using antibodies to putative markers and novel monoclonal antibodies (i.e., ED 18/19/21 and anti-CD205 antibodies) and compare it with a map from mouse counterparts and that of rat thymic dendritic cells. RESULTS Rat TECs were subdivided on the basis of phenotype into three subsets; ED18+ED19+/-keratin 5 (K5)+K8+CD205+ class II MHC (MHCII)+ cortical TECs (cTECs), ED18+ED21-K5-K8+Ulex europaeus lectin 1 (UEA-1)+CD205- medullary TECs (mTEC1s), and ED18+ED21+K5+K8dullUEA-1-CD205- medullary TECs (mTEC2s). Thymic nurse cells were defined in cytosmears as an ED18+ED19+/-K5+K8+ subset of cTECs. mTEC1s preferentially expressed MHCII, claudin-3, claudin-4, and autoimmune regulator (AIRE). Use of ED18 and ED21 antibodies revealed three subsets of TECs in mice as well. We also detected two distinct TEC-free areas in the subcapsular cortex and in the medulla. Rat dendritic cells in the cortex were MHCII+CD103+ but negative for TEC markers, including CD205. Those in the medulla were MHCII+CD103+ and CD205+ cells were found only in the TEC-free area. CONCLUSION Both rats and mice have three TEC subsets with similar phenotypes that can be identified using known markers and new monoclonal antibodies. These findings will facilitate further analysis of TEC subsets and DCs and help to define their roles in thymic selection and in pathological states such as autoimmune disorders.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Cells, Cultured
- Claudin-3/immunology
- Claudin-3/metabolism
- Claudin-4/immunology
- Claudin-4/metabolism
- Epithelial Cells/cytology
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Keratin-5/immunology
- Keratin-5/metabolism
- Keratin-8/immunology
- Keratin-8/metabolism
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Minor Histocompatibility Antigens
- Phenotype
- Plant Lectins/immunology
- Plant Lectins/metabolism
- Rats
- Rats, Inbred Lew
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Thymus Gland/cytology
Collapse
Affiliation(s)
- Yasushi Sawanobori
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Hiashi Ueta
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Christine D. Dijkstra
- Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, Netherlands
| | - Chae Gyu Park
- Laboratory of Immunology, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Motoyasu Satou
- Department of Biochemistry, Dokkyo Medical University, Tochigi, Japan
| | - Yusuke Kitazawa
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Kenjiro Matsuno
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
- * E-mail:
| |
Collapse
|
18
|
Giménez-Barcons M, Casteràs A, Armengol MDP, Porta E, Correa PA, Marín A, Pujol-Borrell R, Colobran R. Autoimmune predisposition in Down syndrome may result from a partial central tolerance failure due to insufficient intrathymic expression of AIRE and peripheral antigens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:3872-9. [PMID: 25217160 DOI: 10.4049/jimmunol.1400223] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Down syndrome (DS), or trisomy of chromosome 21, is the most common genetic disorder associated with autoimmune diseases. Autoimmune regulator protein (AIRE), a transcription factor located on chromosome 21, plays a crucial role in autoimmunity by regulating promiscuous gene expression (pGE). To investigate if autoimmunity in DS is promoted by the reduction of pGE owing to dysregulation of AIRE, we assessed the expression of AIRE and of several peripheral tissue-restricted Ag genes by quantitative PCR in thymus samples from 19 DS subjects and 21 euploid controls. Strikingly, despite the 21 trisomy, AIRE expression was significantly reduced by 2-fold in DS thymuses compared with controls, which was also confirmed by fluorescent microscopy. Allele-specific quantification of intrathymic AIRE showed that despite its lower expression, the three copies are expressed. More importantly, decreased expression of AIRE was accompanied by a reduction of pGE because expression of tissue-restricted Ags, CHRNA1, GAD1, PLP1, KLK3, SAG, TG, and TSHR, was reduced. Of interest, thyroid dysfunction (10 cases of hypothyroidism and 1 of Graves disease) developed in 11 of 19 (57.9%) of the DS individuals and in none of the 21 controls. The thymuses of these DS individuals contained significantly lower levels of AIRE and thyroglobulin, to which tolerance is typically lost in autoimmune thyroiditis leading to hypothyroidism. Our findings provide strong evidence for the fundamental role of AIRE and pGE, namely, central tolerance, in the predisposition to autoimmunity of DS individuals.
Collapse
Affiliation(s)
- Mireia Giménez-Barcons
- Divisió d'Immunologia, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Barcelona 08035, Spain
| | - Anna Casteràs
- Divisió d'Endocrinologia, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Barcelona 08035, Spain
| | - Maria del Pilar Armengol
- Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona 08916, Spain; and
| | - Eduard Porta
- Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona 08916, Spain; and
| | - Paula A Correa
- Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona 08916, Spain; and
| | - Ana Marín
- Divisió d'Immunologia, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Barcelona 08035, Spain
| | - Ricardo Pujol-Borrell
- Divisió d'Immunologia, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Barcelona 08035, Spain; Departament de Biologia Cellular, de Fisiologia i d'Immunologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Roger Colobran
- Divisió d'Immunologia, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Barcelona 08035, Spain; Departament de Biologia Cellular, de Fisiologia i d'Immunologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| |
Collapse
|
19
|
Nishikawa Y, Nishijima H, Matsumoto M, Morimoto J, Hirota F, Takahashi S, Luche H, Fehling HJ, Mouri Y, Matsumoto M. Temporal lineage tracing of Aire-expressing cells reveals a requirement for Aire in their maturation program. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:2585-92. [PMID: 24516201 DOI: 10.4049/jimmunol.1302786] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Understanding the cellular dynamics of Aire-expressing lineage(s) among medullary thymic epithelial cells (AEL-mTECs) is essential for gaining insight into the roles of Aire in establishment of self-tolerance. In this study, we monitored the maturation program of AEL-mTECs by temporal lineage tracing, in which bacterial artificial chromosome transgenic mice expressing tamoxifen-inducible Cre recombinase under control of the Aire regulatory element were crossed with reporter strains. We estimated that the half-life of AEL-mTECs subsequent to Aire expression was ∼7-8 d, which was much longer than that reported previously, owing to the existence of a post-Aire stage. We found that loss of Aire did not alter the overall lifespan of AEL-mTECs, inconsistent with the previous notion that Aire expression in medullary thymic epithelial cells (mTECs) might result in their apoptosis for efficient cross-presentation of self-antigens expressed by AEL-mTECs. In contrast, Aire was required for the full maturation program of AEL-mTECs, as exemplified by the lack of physiological downregulation of CD80 during the post-Aire stage in Aire-deficient mice, thus accounting for the abnormally increased CD80(high) mTECs seen in such mice. Of interest, increased CD80(high) mTECs in Aire-deficient mice were not mTEC autonomous and were dependent on cross-talk with thymocytes. These results further support the roles of Aire in the differentiation program of AEL-mTECs.
Collapse
Affiliation(s)
- Yumiko Nishikawa
- Division of Molecular Immunology, Institute for Enzyme Research, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Thymic epithelial cell development and its dysfunction in human diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:206929. [PMID: 24672784 PMCID: PMC3929497 DOI: 10.1155/2014/206929] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 11/28/2013] [Indexed: 12/01/2022]
Abstract
Thymic epithelial cells (TECs) are the key components in thymic microenvironment for T cells development. TECs, composed of cortical and medullary TECs, are derived from a common bipotent progenitor and undergo a stepwise development controlled by multiple levels of signals to be functionally mature for supporting thymocyte development. Tumor necrosis factor receptor (TNFR) family members including the receptor activator for NFκB (RANK), CD40, and lymphotoxin β receptor (LTβR) cooperatively control the thymic medullary microenvironment and self-tolerance establishment. In addition, fibroblast growth factors (FGFs), Wnt, and Notch signals are essential for establishment of functional thymic microenvironment. Transcription factors Foxn1 and autoimmune regulator (Aire) are powerful modulators of TEC development, differentiation, and self-tolerance. Dysfunction in thymic microenvironment including defects of TEC and thymocyte development would cause physiological disorders such as tumor, infectious diseases, and autoimmune diseases. In the present review, we will summarize our current understanding on TEC development and the underlying molecular signals pathways and the involvement of thymus dysfunction in human diseases.
Collapse
|
21
|
Perniola R, Musco G. The biophysical and biochemical properties of the autoimmune regulator (AIRE) protein. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:326-37. [PMID: 24275490 DOI: 10.1016/j.bbadis.2013.11.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 11/11/2013] [Accepted: 11/18/2013] [Indexed: 01/20/2023]
Abstract
AIRE (for autoimmune regulator) is a multidomain protein that performs a fundamental function in the thymus and possibly in the secondary lymphoid organs: the regulation, especially in the sense of activation, of the process of gene transcription in cell lines deputed to the presentation of self-antigens to the maturing T lymphocytes. The apoptosis of the elements bearing T-cell receptors with critical affinity for the exhibited self-antigens prevents the escape of autoreactive clones and represents a simple and efficient mechanism of deletional self-tolerance. However, AIRE action relies on an articulated complex of biophysical and biochemical properties, in most cases attributable to single subspecialized domains. Here a thorough review of the matter is presented, with a privileged look at the pathogenic changes of AIRE that interfere with such properties and lead to the impairment in its chief function.
Collapse
Affiliation(s)
- Roberto Perniola
- Department of Pediatrics - Neonatal Intensive Care, V. Fazzi Regional Hospital, Piazza F. Muratore, I-73100, Lecce, Italy.
| | - Giovanna Musco
- Biomolecular NMR Laboratory, Center of Translational Genomics and Bioinformatics, Dulbecco Telethon Institute at San Raffaele Scientific Institute, Via Olgettina 58, I-20132, Milan, Italy.
| |
Collapse
|
22
|
Danzl NM, Jeong S, Choi Y, Alexandropoulos K. Identification of novel thymic epithelial cell subsets whose differentiation is regulated by RANKL and Traf6. PLoS One 2014; 9:e86129. [PMID: 24465914 PMCID: PMC3897650 DOI: 10.1371/journal.pone.0086129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 12/10/2013] [Indexed: 12/31/2022] Open
Abstract
Thymic epithelial cells (TECs) are critical for the normal development and function of the thymus. Here, we examined the developmental stages of TECs using quantitative assessment of the cortical and medullary markers Keratin 5 and Keratin 8 (K5 and K8) respectively, in normal and gain/loss of function mutant animals. Gain of function mice overexpressed RANKL in T cells, whereas loss of function animals lacked expression of Traf6 in TECs (Traf6ΔTEC). Assessment of K5 and K8 expression in conjunction with other TEC markers in wild type mice identified novel cortical and medullary TEC populations, expressing different combinations of these markers. RANKL overexpression led to expansion of all medullary TECs (mTECs) and enlargement of the thymic medulla. This in turn associated with a block in thymocyte development and loss of CD4+ CD8+, CD4+ and CD8+ thymocytes. In contrast, Traf6 deletion inhibited the production of most TEC populations including cortical TECs (cTECs), defined by absence of UEA-1 binding and LY51 expression, but had no apparent effect on thymocyte development. These results reveal a large degree of heterogeneity within the TEC compartment and the existence of several populations exhibiting concomitant expression of cortical, medullary and epithelial markers and whose production is regulated by RANKL and Traf6.
Collapse
Affiliation(s)
- Nichole M. Danzl
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Seihwan Jeong
- Department of Medicine, Division of Clinical Immunology, The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Konstantina Alexandropoulos
- Department of Medicine, Division of Clinical Immunology, The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
23
|
Laan M, Peterson P. The many faces of aire in central tolerance. Front Immunol 2013; 4:326. [PMID: 24130560 PMCID: PMC3795325 DOI: 10.3389/fimmu.2013.00326] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/24/2013] [Indexed: 11/13/2022] Open
Abstract
Although the role that Autoimmune Regulator (Aire) plays in the induction of central tolerance is well known, the precise cellular and molecular mechanisms are still unclear and debated. In the prevailing view, Aire serves mainly as a direct inducer of tissue-specific antigens. However, there is a growing amount of evidence suggesting that Aire modulates the differentiation program of medullary thymic epithelial cells, which may directly contribute to the negative selection of self-reactive thymocytes. In addition, Aire has been shown to regulate the expression of many intrathymic chemokines that are required for the proper localization of thymocytes and dendritic cells, and thus are potentially important for direct and indirect self-antigen presentation in the thymic medulla. Further, recent evidence suggests that the induction of certain antigen-specific regulatory T-cells that translocate to tumors and peripheral tissues can be Aire dependent and may contribute to tissue-specific tolerance. This review summarizes the current understanding of the effects of Aire on these alternative mechanisms for the induction of Aire-induced central tolerance.
Collapse
Affiliation(s)
- Martti Laan
- Molecular Pathology, Faculty of Medicine, Institute of Biomedicine and Translational Medicine, Tartu University , Tartu , Estonia
| | | |
Collapse
|
24
|
Abstract
Loss-of-function mutations in the Autoimmune Regulator (AIRE) gene cause a rare inherited form of autoimmune disease, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, also known as autoimmune polyglandular syndrome type 1. The patients suffer from multiple endocrine deficiencies, the most common manifestations being hypoparathyroidism, Addison’s disease, hypogonadism, and secondary amenorrhea, usually accompanied by typical autoantibodies against the target tissues. Chronic mucocutaneous candidiasis is also a prominent part of the disease. The highest expression of AIRE is found in medullary thymic epithelial cells (mTECs). Murine studies suggest that it promotes ectopic transcription of self antigens in mTECs and is thus important for negative selection. However, failed negative selection alone is not enough to explain key findings in human patients, necessitating the search for alternative or additional pathogenetic mechanisms. A striking feature of the human AIRE-deficient phenotype is that all patients develop high titers of neutralizing autoantibodies against type I interferons, which have been shown to downregulate the expression of interferon-controlled genes. These autoantibodies often precede clinical symptoms and other autoantibodies, suggesting that they are a reflection of the pathogenetic process. Other cytokines are targeted as well, notably those produced by Th17 cells; these autoantibodies have been linked to the defect in anti-candida defenses. A defect in regulatory T cells has also been reported in several studies and seems to affect already the recent thymic emigrant population. Taken together, these findings in human patients point to a widespread disruption of T cell development and regulation, which is likely to have its origins in an abnormal thymic milieu. The absence of functional AIRE in peripheral lymphoid tissues may also contribute to the pathogenesis of the disease.
Collapse
Affiliation(s)
- T Petteri Arstila
- Department of Bacteriology and Immunology, Immunobiology Research Program, Haartman Institute, University of Helsinki , Helsinki , Finland
| | | |
Collapse
|
25
|
Garfin PM, Min D, Bryson JL, Serwold T, Edris B, Blackburn CC, Richie ER, Weinberg KI, Manley NR, Sage J, Viatour P. Inactivation of the RB family prevents thymus involution and promotes thymic function by direct control of Foxn1 expression. ACTA ACUST UNITED AC 2013; 210:1087-97. [PMID: 23669396 PMCID: PMC3674705 DOI: 10.1084/jem.20121716] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
RB family genes control T cell production and promote thymic involution through reducing Foxn1 expression in thymic epithelial cells. Thymic involution during aging is a major cause of decreased production of T cells and reduced immunity. Here we show that inactivation of Rb family genes in young mice prevents thymic involution and results in an enlarged thymus competent for increased production of naive T cells. This phenotype originates from the expansion of functional thymic epithelial cells (TECs). In RB family mutant TECs, increased activity of E2F transcription factors drives increased expression of Foxn1, a central regulator of the thymic epithelium. Increased Foxn1 expression is required for the thymic expansion observed in Rb family mutant mice. Thus, the RB family promotes thymic involution and controls T cell production via a bone marrow–independent mechanism, identifying a novel pathway to target to increase thymic function in patients.
Collapse
Affiliation(s)
- Phillip M Garfin
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sun L, Luo H, Li H, Zhao Y. Thymic epithelial cell development and differentiation: cellular and molecular regulation. Protein Cell 2013; 4:342-55. [PMID: 23589020 DOI: 10.1007/s13238-013-3014-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/11/2013] [Indexed: 11/26/2022] Open
Abstract
Thymic epithelial cells (TECs) are one of the most important components in thymic microenvironment supporting thymocyte development and maturation. TECs, composed of cortical and medullary TECs, are derived from a common bipotent progenitor, mediating thymocyte positive and negative selections. Multiple levels of signals including intracellular signaling networks and cell-cell interaction are required for TEC development and differentiation. Transcription factors Foxn1 and autoimmune regulator (Aire) are powerful regulators promoting TEC development and differentiation. Crosstalks with thymocytes and other stromal cells for extrinsic signals like RANKL, CD40L, lymphotoxin, fibroblast growth factor (FGF) and Wnt are also definitely required to establish a functional thymic microenvironment. In this review, we will summarize our current understanding about TEC development and differentiation, and its underlying multiple signal pathways.
Collapse
Affiliation(s)
- Lina Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | |
Collapse
|
27
|
Otero DC, Baker DP, David M. IRF7-dependent IFN-β production in response to RANKL promotes medullary thymic epithelial cell development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:3289-98. [PMID: 23440417 PMCID: PMC3608802 DOI: 10.4049/jimmunol.1203086] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The contributions of IFN regulatory factor (IRF) 3/7 and the type I IFNs IFN-α/β to the innate host defense have been extensively investigated; however, their role in thymic development is less clear. In this study, we show that mice lacking the type I IFN receptor IFN-α/β receptor (IFNAR) or the downstream transcription factor STAT1 harbor a significant reduction in self-Ag-presenting, autoimmune regulator (AIRE)(+) medullary thymic epithelial cells (mTECs). Constitutive IFNAR signaling occurs in the thymic medulla in the absence of infection or inflammation. Receptor activator for NF-κB (RANK) ligand stimulation results in IFN-β upregulation, which in turn inhibits RANK signaling and facilitates AIRE expression in mTECs. Finally, we find that IRF7 is required for thymic IFN-β induction, maintenance of thymic architecture, and mTEC differentiation. We conclude that spatially and temporally coordinated cross talks between the RANK ligand/RANK and IRF7/IFN-β/IFNAR/STAT1 pathways are essential for differentiation of AIRE(+) mTECs.
Collapse
Affiliation(s)
- Dennis C. Otero
- Division of Biological Sciences and UCSD Cancer Center, University of California San Diego, La Jolla, CA, USA
| | | | - Michael David
- Division of Biological Sciences and UCSD Cancer Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
28
|
Pinto S, Schmidt K, Egle S, Stark HJ, Boukamp P, Kyewski B. An organotypic coculture model supporting proliferation and differentiation of medullary thymic epithelial cells and promiscuous gene expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:1085-93. [PMID: 23269248 DOI: 10.4049/jimmunol.1201843] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Understanding intrathymic T cell differentiation has been greatly aided by the development of various reductionist in vitro models that mimic certain steps/microenvironments of this complex process. Most models focused on the faithful in vitro restoration of T cell differentiation and selection. In contrast, suitable in vitro models emulating the developmental pathways of the two major thymic epithelial cell lineages--cortical thymic epithelial cells and medullary thymic epithelial cells (mTECs)--are yet to be developed. In this regard, lack of an in vitro model mimicking the developmental biology of the mTEC lineage has hampered the molecular analysis of the so-called "promiscuous expression" of tissue-restricted genes, a key property of terminally differentiated mTECs. Based on the close biological relationship between the skin and thymus epithelial cell compartments, we adapted a three-dimensional organotypic coculture model, originally developed to provide a bona fide in vitro dermal equivalent, for the culture of isolated mTECs. This three-dimensional model preserves key features of mTECs: proliferation and terminal differentiation of CD80(lo), Aire(-) mTECs into CD80(hi), Aire(+) mTECs; responsiveness to RANKL; and sustained expression of FoxN1, Aire, and tissue-restricted genes in CD80(hi) mTECs. This in vitro culture model should facilitate the identification of molecular components and pathways involved in mTEC differentiation in general and in promiscuous gene expression in particular.
Collapse
Affiliation(s)
- Sheena Pinto
- Division of Developmental Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
The development of CD4(+) helper and CD8(+) cytotoxic T-cells expressing the αβ form of the T-cell receptor (αβTCR) takes place in the thymus, a primary lymphoid organ containing distinct cortical and medullary microenvironments. While the cortex represents a site of early T-cell precursor development, and the positive selection of CD4(+)8(+) thymocytes, the thymic medulla plays a key role in tolerance induction, ensuring that thymic emigrants are purged of autoreactive αβTCR specificities. In recent years, advances have been made in understanding the development and function of thymic medullary epithelial cells, most notably the subset defined by expression of the Autoimmune Regulator (Aire) gene. Here, we summarize current knowledge of the developmental mechanisms regulating thymus medulla development, and examine the role of the thymus medulla in recessive (negative selection) and dominant (T-regulatory cell) tolerance.
Collapse
|
30
|
Danso-Abeam D, Staats KA, Franckaert D, Van Den Bosch L, Liston A, Gray DHD, Dooley J. Aire mediates thymic expression and tolerance of pancreatic antigens via an unconventional transcriptional mechanism. Eur J Immunol 2013; 43:75-84. [PMID: 23041971 DOI: 10.1002/eji.201242761] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/24/2012] [Accepted: 10/01/2012] [Indexed: 01/20/2023]
Abstract
The autoimmune regulator (Aire), mediates central tolerance of peripheral self. Its activity in thymic epithelial cells (TECs) directs the ectopic expression of thousands of tissue-restricted antigens (TRAs), causing the deletion of autoreactive thymocytes. The molecular mechanisms orchestrating the breadth of transcriptional regulation by Aire remain unknown. One prominent model capable of explaining both the uniquely high number of Aire-dependent targets and their specificity posits that tissue-specific transcription factors induced by Aire directly activate their canonical targets, exponentially adding to the total number of Aire-dependent TRAs. To test this "Hierarchical Transcription" model, we analysed mice deficient in the pancreatic master transcription factor pancreatic and duodenal homeobox 1 (Pdx1), specifically in TECs (Pdx1(ΔFoxn1) ), for the expression and tolerance of pancreatic TRAs. Surprisingly, we found that lack of Pdx1 in TECs did not reduce the transcription of insulin or somatostatin, or alter glucagon expression. Moreover, in a model of thymic deletion driven by a neo-TRA under the control of the insulin promoter, Pdx1 in TECs was not required to affect thymocyte deletion or the generation of regulatory T (Treg) cells. These findings suggest that the capacity of Aire to regulate expression of a huge array of TRAs relies solely on an unconventional transcriptional mechanism, without intermediary transcription factors.
Collapse
|
31
|
Wu G, Hirabayashi K, Sato S, Akiyama N, Akiyama T, Shiota K, Yagi S. DNA methylation profile of Aire-deficient mouse medullary thymic epithelial cells. BMC Immunol 2012; 13:58. [PMID: 23116172 PMCID: PMC3546423 DOI: 10.1186/1471-2172-13-58] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 10/27/2012] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Medullary thymic epithelial cells (mTECs) are characterized by ectopic expression of self-antigens during the establishment of central tolerance. The autoimmune regulator (Aire), which is specifically expressed in mTECs, is responsible for the expression of a large repertoire of tissue-restricted antigens (TRAs) and plays a role in the development of mTECs. However, Aire-deficient mTECs still express TRAs. Moreover, a subset of mTECs, which are considered to be at a stage of terminal differentiation, exists in the Aire-deficient thymus. The phenotype of a specific cell type in a multicellular organism is governed by the epigenetic regulation system. DNA methylation modification is an important component of this system. Every cell or tissue type displays a DNA methylation profile, consisting of tissue-dependent and differentially methylated regions (T-DMRs), and this profile is involved in cell-type-specific genome usage. The aim of this study was to examine the DNA methylation profile of mTECs by using Aire-deficient mTECs as a model. RESULTS We identified the T-DMRs of mTECs (mTEC-T-DMRs) via genome-wide DNA methylation analysis of Aire(-/-) mTECs by comparison with the liver, brain, thymus, and embryonic stem cells. The hypomethylated mTEC-T-DMRs in Aire(-/-) mTECs were associated with mTEC-specific genes, including Aire, CD80, and Trp63, as well as other genes involved in the RANK signaling pathway. While these mTEC-T-DMRs were also hypomethylated in Aire(+/+) mTECs, they were hypermethylated in control thymic stromal cells. We compared the pattern of DNA methylation levels at a total of 55 mTEC-T-DMRs and adjacent regions and found that the DNA methylation status was similar for Aire(+/+) and Aire(-/-) mTECs but distinct from that of athymic cells and tissues. CONCLUSIONS These results indicate a unique DNA methylation profile that is independent of Aire in mTECs. This profile is distinct from other cell types in the thymic microenvironment and is indicated to be involved in the differentiation of the mTEC lineage.
Collapse
Affiliation(s)
- Guoying Wu
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences /Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Keiji Hirabayashi
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences /Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Shinya Sato
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences /Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Nobuko Akiyama
- Division of Cellular and Molecular Biology, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokane-dai, Minato-ku, Tokyo, 108-8639, Japan
| | - Taishin Akiyama
- Division of Cellular and Molecular Biology, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokane-dai, Minato-ku, Tokyo, 108-8639, Japan
| | - Kunio Shiota
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences /Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Shintaro Yagi
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences /Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| |
Collapse
|
32
|
Akiyama T, Shinzawa M, Akiyama N. TNF receptor family signaling in the development and functions of medullary thymic epithelial cells. Front Immunol 2012; 3:278. [PMID: 22969770 PMCID: PMC3432834 DOI: 10.3389/fimmu.2012.00278] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 08/16/2012] [Indexed: 11/25/2022] Open
Abstract
Thymic epithelial cells (TECs) provide the microenvironment required for the development of T cells in the thymus. A unique property of medullary thymic epithelial cells (mTECs) is their expression of a wide range of tissue-restricted self-antigens, critically regulated by the nuclear protein AIRE, which contributes to the selection of the self-tolerant T cell repertoire, thereby suppressing the onset of autoimmune diseases. The TNF receptor family (TNFRF) protein receptor activator of NF-κB (RANK), CD40 and lymphotoxin β receptor (LtβR) regulate the development and functions of mTECs. The engagement of these receptors with their specific ligands results in the activation of the NF-κB family of transcription factors. Two NF-κB activation pathways, the classical and non-classical pathways, promote the development of mature mTECs induced by these receptors. Consistently, TNF receptor-associated factor (TRAF6), the signal transducer of the classical pathway, and NF-κB inducing kinase (NIK), the signal transducer of the non-classical pathway, are essential for the development of mature mTECs. This review summarizes the current understanding of how the signaling by the TNF receptor family controls the development and functions of mTEC.
Collapse
Affiliation(s)
- Taishin Akiyama
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo Tokyo, Japan
| | | | | |
Collapse
|
33
|
Seach N, Wong K, Hammett M, Boyd RL, Chidgey AP. Purified enzymes improve isolation and characterization of the adult thymic epithelium. J Immunol Methods 2012; 385:23-34. [PMID: 22910002 DOI: 10.1016/j.jim.2012.07.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Revised: 07/28/2012] [Accepted: 07/31/2012] [Indexed: 11/30/2022]
Abstract
The reproducible isolation and accurate characterization of thymic epithelial cell (TEC) subsets is of critical importance to the ongoing study of thymopoiesis and its functional decline with age. The study of adult TEC, however, is significantly hampered due to the severely low stromal to hematopoietic cell ratio. Non-biased digestion and enrichment protocols are thus essential to ensure optimal cell yield and accurate representation of stromal subsets, as close as possible to their in vivo representation. Current digestion protocols predominantly involve diverse, relatively impure enzymatic variants of crude collagenase and collagenase/dispase (col/disp) preparations, which have variable efficacy and are often suboptimal in their ability to mediate complete digestion of thymus tissue. To address these issues we compared traditional col/disp preparations with the latest panel of Liberase products that contain a blend of highly purified collagenase and neutral protease enzymes. Liberase enzymes revealed a more rapid, complete dissociation of thymus tissue; minimizing loss of viability and increasing recovery of thymic stromal cell (TSC) elements. In particular, the recovery and viability of TEC, notably the rare cortical subsets, were significantly enhanced with Liberase products containing medium to high levels of thermolysin. The improved stromal dissociation led to numerically increased TEC yield and total TEC RNA isolated from pooled digests of adult thymus. Furthermore, the increased recovery of TEC enhanced resolution and quantification of TEC subsets in both adult and aged mice, facilitating flow cytometric analysis on a per thymus basis. We further refined the adult TEC phenotype by correlating surface expression of known TEC markers, with expression of intracellular epithelial lineage markers, Keratin 5 and Keratin 8. The data reveal more extensive expression of K8 than previously recognized and indicates considerable heterogeneity still exists within currently defined adult TEC subsets.
Collapse
Affiliation(s)
- Natalie Seach
- Monash Immunology and Stem Cell Laboratories, Level 3, STRIP-1, Building 75, Monash University, Wellington Rd. Clayton, Victoria 3800, Australia
| | | | | | | | | |
Collapse
|
34
|
Liiv I, Haljasorg U, Kisand K, Maslovskaja J, Laan M, Peterson P. AIRE-induced apoptosis is associated with nuclear translocation of stress sensor protein GAPDH. Biochem Biophys Res Commun 2012; 423:32-7. [PMID: 22613203 DOI: 10.1016/j.bbrc.2012.05.057] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 01/19/2023]
Abstract
AIRE (Autoimmune Regulator) has a central role in the transcriptional regulation of self-antigens in medullary thymic epithelial cells, which is necessary for negative selection of autoreactive T cells. Recent data have shown that AIRE can also induce apoptosis, which may be linked to cross-presentation of these self-antigens. Here we studied AIRE-induced apoptosis using AIRE over-expression in a thymic epithelial cell line as well as doxycycline-inducible HEK293 cells. We show that the HSR/CARD domain in AIRE together with a nuclear localization signal is sufficient to induce apoptosis. In the nuclei of AIRE-positive cells, we also found an increased accumulation of a glycolytic enzyme, glyceraldehyde-3-phosphate (GAPDH) reflecting cellular stress and apoptosis. Additionally, AIRE-induced apoptosis was inhibited with an anti-apoptotic agent deprenyl that blocks GAPDH nitrosylation and nuclear translocation. We propose that the AIRE-induced apoptosis pathway is associated with GAPDH nuclear translocation and induction of NO-induced cellular stress in AIRE-expressing cells.
Collapse
Affiliation(s)
- Ingrid Liiv
- Molecular Pathology, Institute of General and Molecular Pathology, University of Tartu, Tartu, Estonia.
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Somatic recombination of TCR genes in immature thymocytes results in some cells with useful TCR specificities, but also many with useless or potentially self-reactive specificities. Thus thymic selection mechanisms operate to shape the T-cell repertoire. Thymocytes that have a TCR with low affinity for self-peptide-MHC complexes are positively selected to further differentiate and function in adaptive immunity, whereas useless ones die by neglect. Clonal deletion and clonal diversion (Treg differentiation) are the major processes in the thymus that eliminate or control self-reactive T cells. Although these processes are thought to be efficient, they fail to control self-reactivity in all circumstances. Thus, peripheral tolerance processes exist wherein self-reactive T cells become functionally unresponsive (anergy) or are deleted after encountering self-antigens outside of the thymus. Recent advances in mechanistic studies of central and peripheral T-cell tolerance are promoting the development of therapeutic strategies to treat autoimmune disease and cancer and improve transplantation outcome.
Collapse
Affiliation(s)
- Yan Xing
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, 55455, USA
| | | |
Collapse
|
36
|
Wang X, Laan M, Bichele R, Kisand K, Scott HS, Peterson P. Post-Aire maturation of thymic medullary epithelial cells involves selective expression of keratinocyte-specific autoantigens. Front Immunol 2012; 3:19. [PMID: 22448160 PMCID: PMC3310317 DOI: 10.3389/fimmu.2012.00019] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The autoimmune regulator (Aire)-directed ectopic expression of tissue-specific antigens (TSAs) by mature medullary thymic epithelial cells (mTECs) has been viewed as an essential mechanism in the induction of central tolerance. Recent data suggest that the survival of mTECs extends beyond the Aire+ cell population to form the post-Aire mTEC population and Hassall’s corpuscles (HCs). The nature and function of these post-Aire epithelial cells and structures, however, have remained unidentified. In this study, we characterized in detail the end-stage development of mTECs and HCs in both Aire-sufficient and Aire-deficient mice. In addition, using a transgenic mouse model in which the LacZ reporter gene is under the control of the endogenous Aire promoter, we purified and analyzed the post-Aire mTECs to characterize their function. We showed that the end-stage maturation of mTECs closely resembles that of keratinocytes and that the lack of Aire results in a marked block of mTEC differentiation, which is partially overcome by ligands for RANK and CD40. We also provide evidence that, during mTEC development, Aire is expressed only once and during a limited 1–2 day period. The following loss of Aire expression is accompanied by a quick downregulation of MHC class II and CD80, and of most of the Aire-dependent and Aire-independent TSAs, with the exception of keratinocyte-specific genes. In the final stage of maturation, the mTECs lose their nuclei to become HCs and specifically express desmogleins (DGs) 1 and 3, which, via cross-presentation by APCs, may contribute to tolerance against these pemphigus vulgaris-related TSAs.
Collapse
Affiliation(s)
- Xiaoping Wang
- Molecular Pathology, Institute of General and Molecular Pathology, University of Tartu, Tartu, Estonia
| | | | | | | | | | | |
Collapse
|
37
|
The thymic epithelial microRNA network elevates the threshold for infection-associated thymic involution via miR-29a mediated suppression of the IFN-α receptor. Nat Immunol 2011; 13:181-7. [PMID: 22179202 DOI: 10.1038/ni.2193] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 11/21/2011] [Indexed: 11/08/2022]
Abstract
Thymic output is a dynamic process, with high activity at birth punctuated by transient periods of involution during infection. Interferon-α (IFN-α) is a critical molecular mediator of pathogen-induced thymic involution, yet despite the importance of thymic involution, relatively little is known about the molecular integrators that establish sensitivity. Here we found that the microRNA network dependent on the endoribonuclease Dicer, and specifically microRNA miR-29a, was critical for diminishing the sensitivity of the thymic epithelium to simulated infection signals, protecting the thymus against inappropriate involution. In the absence of Dicer or the miR-29a cluster in the thymic epithelium, expression of the IFN-α receptor by the thymic epithelium was higher, which allowed suboptimal signals to trigger rapid loss of thymic cellularity.
Collapse
|
38
|
Danso-Abeam D, Humblet-Baron S, Dooley J, Liston A. Models of aire-dependent gene regulation for thymic negative selection. Front Immunol 2011; 2:14. [PMID: 22566805 PMCID: PMC3342030 DOI: 10.3389/fimmu.2011.00014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Accepted: 04/21/2011] [Indexed: 11/13/2022] Open
Abstract
Mutations in the autoimmune regulator (AIRE) gene lead to autoimmune polyendocrinopathy syndrome type 1 (APS1), characterized by the development of multi-organ autoimmune damage. The mechanism by which defects in AIRE result in autoimmunity has been the subject of intense scrutiny. At the cellular level, the working model explains most of the clinical and immunological characteristics of APS1, with AIRE driving the expression of tissue-restricted antigens (TRAs) in the epithelial cells of the thymic medulla. This TRA expression results in effective negative selection of TRA-reactive thymocytes, preventing autoimmune disease. At the molecular level, the mechanism by which AIRE initiates TRA expression in the thymic medulla remains unclear. Multiple different models for the molecular mechanism have been proposed, ranging from classical transcriptional activity, to random induction of gene expression, to epigenetic tag recognition effect, to altered cell biology. In this review, we evaluate each of these models and discuss their relative strengths and weaknesses.
Collapse
|
39
|
Abstract
In the thymus, developing T cells that react against self-antigens with high affinity are deleted in the process of negative selection. An essential component of this process is the display of self-antigens, including those whose expression are usually restricted to specific tissues, to developing T cells within the thymus. The Autoimmune Regulator (Aire) gene plays a crucial role in the expression of tissue specific self-antigens within the thymus, and disruption of Aire function results in spontaneous autoimmunity in both humans and mice. Recent advances have been made in our understanding of how Aire influences the expression of thousands of tissue-specific antigens in the thymus. Additional roles of Aire, including roles in chemokine and cytokine expression, have also been revealed. Factors important in the differentiation of Aire-expressing medullary thymic epithelial cells have been defined. Finally, the identity of antigen presenting cells in negative selection, including the role of medullary thymic epithelial cells in displaying tissue specific antigens to T cells, has also been clarified.
Collapse
Affiliation(s)
- Mark S. Anderson
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Maureen A. Su
- Inflammatory Diseases Institute and Department of Pediatrics, University of North Carolina, Chapel Hill, Chapel Hill, NC
| |
Collapse
|
40
|
McLelland BT, Gravano D, Castillo J, Montoy S, Manilay JO. Enhanced isolation of adult thymic epithelial cell subsets for multiparameter flow cytometry and gene expression analysis. J Immunol Methods 2011; 367:85-94. [PMID: 21354161 DOI: 10.1016/j.jim.2011.02.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 12/13/2010] [Accepted: 02/18/2011] [Indexed: 12/17/2022]
Abstract
The epithelial cells (TECs) are microenvironmental niche cells which support T lymphocyte development in the thymus. Most studies of TEC biology have focused on TEC at the fetal stage of development, whereas the biology of adult-stage TECs is not as well-understood. Delineating the molecular mechanisms that control adult TEC differentiation has implications for the success of T-lymphocyte based therapies for autoimmune diseases and induction of immunological tolerance to stem cell-derived tissues. Detailed analysis of adult TECs is technically challenging due to their rarity, their diminishing numbers with age, and the limited number of markers to distinguish between unique TEC subpopulations. Here, we have devised an improved isolation protocol for adult mouse TECs and combined it with six-color multiparameter flow cytometry. Using these techniques, we have identified four distinct subsets of CD45- EpCAM+ TECs in adult mice: a) UEA1(low) CDR1(low) (UC(low)); b) UEA1(high) CDR1(high)(UC(high)); c) UEA1(low) CDR1(high) MHC(high) (cTEC); and d) UEA1(high)CDR1(low) MHC(int/high) (mTEC). PCR analysis verified that these TEC subsets differentially expressed known TEC genes. TEC subsets were further analyzed using high-throughput quantitative PCR arrays to reveal novel genes that could be important for TEC subset maintenance. Intracellular staining for keratin-5 and keratin-8 can also be added, but our results suggest that keratin expression alone cannot be used to distinguish adult TEC subsets. Our enhanced isolation allows for detailed analysis of rare TEC subpopulations in the adult mouse at the cellular and molecular levels.
Collapse
Affiliation(s)
- Bryce T McLelland
- School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, CA 95340, USA
| | | | | | | | | |
Collapse
|
41
|
Matsumoto M. Contrasting models for the roles of Aire in the differentiation program of epithelial cells in the thymic medulla. Eur J Immunol 2011; 41:12-7. [PMID: 21182071 DOI: 10.1002/eji.201041024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The current prevailing view regarding the role of Aire in self-tolerance is that it is involved in the transcriptional control of many tissue-restricted self-antigen genes in thymic epithelial cells in the medulla (mTECs); however, accumulating evidence also suggests that Aire has other roles, e.g. in mTEC differentiation, and furthermore that Aire can either promote or inhibit the mTEC differentiation program, i.e. Aire does not play a neutral role in mTEC differentiation. This review discusses when and how Aire plays an important role in controlling the organization of mTECs required for the expression of self-antigen genes.
Collapse
Affiliation(s)
- Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, University of Tokushima, Tokushima, Japan.
| |
Collapse
|
42
|
Abstract
The development of regulatory T (Treg) cells is essential for the maintenance of immune tolerance and homeostasis. Here, we review recent studies that have advanced our understanding of Treg cell differentiation. In the thymus, TCR specificity to self-antigen appears to be a primary determinant for Treg cell lineage commitment, with c-Rel being an important factor that links T cell receptor (TCR) engagement and Foxp3 expression, along with cytokines and costimulatory molecules. It is also clear that postthymic events shape the peripheral Treg cell population. This includes preferential maintenance of Treg cells specific to self-antigens presented in the periphery, as well as the de novo generation of Treg cells from conventional Foxp3(-) T cells. The process of peripheral Treg cell differentiation shares some features with thymic Treg cell development, but there are notable differences. Together, thymic and peripheral Treg cell differentiation appear to generate an "imprint" of both self- and foreign antigens in the peripheral Treg cell population to provide dominant tolerance.
Collapse
Affiliation(s)
- Hyang-Mi Lee
- Department of Internal Medicine, Division of Rheumatology, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | |
Collapse
|
43
|
Nitta T, Ohigashi I, Nakagawa Y, Takahama Y. Cytokine crosstalk for thymic medulla formation. Curr Opin Immunol 2010; 23:190-7. [PMID: 21194915 DOI: 10.1016/j.coi.2010.12.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 11/24/2010] [Accepted: 12/03/2010] [Indexed: 01/12/2023]
Abstract
The medullary microenvironment of the thymus plays a crucial role in the establishment of self-tolerance through the deletion of self-reactive thymocytes and the generation of regulatory T cells. Crosstalk or bidirectional signal exchanges between developing thymocytes and medullary thymic epithelial cells (mTECs) contribute to the formation of the thymic medulla. Recent studies have identified the molecules that mediate thymic crosstalk. Tumor necrosis factor superfamily cytokines, including RANKL, CD40L, and lymphotoxin, produced by positively selected thymocytes and lymphoid tissue inducer cells promote the proliferation and differentiation of mTECs. In return, CCR7 ligand chemokines produced by mTECs facilitate the migration of positively selected thymocytes to the medulla. The cytokine crosstalk between developing thymocytes and mTECs nurtures the formation of the thymic medulla and thereby regulates the establishment of self-tolerance.
Collapse
Affiliation(s)
- Takeshi Nitta
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | |
Collapse
|
44
|
Laakso SM, Laurinolli TT, Rossi LH, Lehtoviita A, Sairanen H, Perheentupa J, Kekäläinen E, Arstila TP. Regulatory T cell defect in APECED patients is associated with loss of naive FOXP3(+) precursors and impaired activated population. J Autoimmun 2010; 35:351-7. [PMID: 20805020 DOI: 10.1016/j.jaut.2010.07.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 07/10/2010] [Accepted: 07/29/2010] [Indexed: 12/11/2022]
Abstract
The pathogenetic mechanisms of organ-specific autoimmune diseases remain obscured by the complexity of the genetic and environmental factors participating in the breakdown of tolerance. A unique opportunity to study the pathogenesis of human autoimmunity is provided by autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), a rare inherited autoimmune disease caused by mutations in Autoimmune Regulator (AIRE) gene. Loss of AIRE function disrupts the deletion of autoreactive T cells and impairs the suppressive function of regulatory T (Treg) cells. Here we show by multiparameter flow cytometry that in healthy controls the peripheral naive Treg cell subset forms a slowly dividing, persistent reservoir of recent thymic emigrants (RTEs). In APECED patients the RTE Treg cells show accelerated turnover and shift to the activated pool and the RTE reservoir is depleted. Moreover, the activated Treg cell population in the patients expresses significantly less Forkhead box protein P3 (FOXP3) than in the healthy controls, consistent with the impairment of peripheral activation. Our results indicate that in addition to their thymic effects, loss-of-function mutations in AIRE disrupt the peripheral homeostasis and activation of Treg cells. This may synergize with failed negative selection to cause APECED.
Collapse
Affiliation(s)
- Sini M Laakso
- Haartman Institute, Department of Immunology, PB21, 00014 University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Guo J, Rahman M, Cheng L, Zhang S, Tvinnereim A, Su DM. Morphogenesis and maintenance of the 3D thymic medulla and prevention of nude skin phenotype require FoxN1 in pre- and post-natal K14 epithelium. J Mol Med (Berl) 2010; 89:263-77. [PMID: 21109991 DOI: 10.1007/s00109-010-0700-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 10/31/2010] [Accepted: 11/05/2010] [Indexed: 12/30/2022]
Abstract
Expansion of thymic epithelial cysts represents disruption of an organized three-dimensional (3D) thymic epithelial cell (TEC) meshwork, which is crucial for T-lymphocyte development. Although the FoxN1-null mutant develops a rudimentary two-dimensional (2D) cystic thymus, 2D thymic cyst lining resulting from a dGUO culture was reported to be FoxN1-independent; thus, it is unclear whether loss of FoxN1 facilitates cyst formation and whether FoxN1 regulates the morphogenesis and maintenance of the 3D thymic microstructure. Using the loxP-floxed-FoxN1 mouse model, we demonstrated that specific deletion of FoxN1 in keratin (K)-14 promoter-driven TECs induced the loss of 3D thymic medullary structure by producing a large number of morphologic pulmonary alveolar-like 2D epithelial cysts, which increased with age. The cystic lining was positive for differential polarized keratins and had strong claudin-3,4, but reduced MHC-II, expression. However, an increased percentage of claudin-3,4(+) TECs, which are presumptive precursors of UEA-1(+) and Aire(+) mature medullary TECs, failed to promote the development of these mature descendants. Meanwhile, the K14Cre-mediated FoxN1 deletion alone was sufficient to induce a complete hair follicle defect, causing a nude phenotype in the skin, but was not sufficient to cause a complete loss of the thymus. All these changes to occur require deletion of FoxN1 in both prenatal (Cre-recombinase from parents during fertilization) and postnatal (Cre-recombinase from offspring themselves after fertilization) life. These findings provide new insights into FoxN1 regulation of 3D thymic epithelial morphogenesis and maintenance, the distinct impacts of FoxN1 in the K14 epithelial subset of the thymus and skin, and its postnatal requirement.
Collapse
Affiliation(s)
- Jianfei Guo
- Department of Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | | | | | | | | | | |
Collapse
|
46
|
Gravano DM, McLelland BT, Horiuchi K, Manilay JO. ADAM17 deletion in thymic epithelial cells alters aire expression without affecting T cell developmental progression. PLoS One 2010; 5:e13528. [PMID: 20976004 PMCID: PMC2958126 DOI: 10.1371/journal.pone.0013528] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 09/20/2010] [Indexed: 12/18/2022] Open
Abstract
Background Cellular interactions between thymocytes and thymic stromal cells are critical for normal T cell development. Thymic epithelial cells (TECs) are important stromal niche cells that provide essential growth factors, cytokines, and present self-antigens to developing thymocytes. The identification of genes that mediate cellular crosstalk in the thymus is ongoing. One candidate gene, Adam17, encodes a metalloprotease that functions by cleaving the ectodomain of several transmembrane proteins and regulates various developmental processes. In conventional Adam17 knockout mice, a non-cell autonomous role for ADAM17 in adult T cell development was reported, which strongly suggested that expression of ADAM17 in TECs was required for normal T cell development. However, knockdown of Adam17 results in multisystem developmental defects and perinatal lethality, which has made study of the role of Adam17 in specific cell types difficult. Here, we examined T cell and thymic epithelial cell development using a conditional knockout approach. Methodology/Principal Findings We generated an Adam17 conditional knockout mouse in which floxed Adam17 is deleted specifically in TECs by Cre recombinase under the control of the Foxn1 promoter. Normal T cell lineage choice and development through the canonical αβ T cell stages was observed. Interestingly, Adam17 deficiency in TECs resulted in reduced expression of the transcription factor Aire. However, no alterations in the patterns of TEC phenotypic marker expression and thymus morphology were noted. Conclusions/Significance In contrast to expectation, our data clearly shows that absence of Adam17 in TECs is dispensable for normal T cell development. Differentiation of TECs is also unaffected by loss of Adam17 based on phenotypic markers. Surprisingly, we have uncovered a novel genetic link between Adam17and Aire expression in vivo. The cell type in which ADAM17 mediates its non-cell autonomous impact and the mechanisms by which it regulates intrathymic T cell development remain to be identified.
Collapse
Affiliation(s)
- David M. Gravano
- School of Natural Sciences, University of California at Merced, Merced, California, United States of America
| | - Bryce T. McLelland
- School of Natural Sciences, University of California at Merced, Merced, California, United States of America
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery and Department of Anti-aging Orthopedic Research, School of Medicine, Keio University, Tokyo, Japan
| | - Jennifer O. Manilay
- School of Natural Sciences, University of California at Merced, Merced, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
White AJ, Nakamura K, Jenkinson WE, Saini M, Sinclair C, Seddon B, Narendran P, Pfeffer K, Nitta T, Takahama Y, Caamano JH, Lane PJL, Jenkinson EJ, Anderson G. Lymphotoxin signals from positively selected thymocytes regulate the terminal differentiation of medullary thymic epithelial cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:4769-76. [PMID: 20861360 PMCID: PMC3826119 DOI: 10.4049/jimmunol.1002151] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The thymic medulla represents a key site for the induction of T cell tolerance. In particular, autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) provide a spectrum of tissue-restricted Ags that, through both direct presentation and cross-presentation by dendritic cells, purge the developing T cell repertoire of autoimmune specificities. Despite this role, the mechanisms of Aire(+) mTEC development remain unclear, particularly those stages that occur post-Aire expression and represent mTEC terminal differentiation. In this study, in mouse thymus, we analyze late-stage mTEC development in relation to the timing and requirements for Aire and involucrin expression, the latter a marker of terminally differentiated epithelium including Hassall's corpuscles. We show that Aire expression and terminal differentiation within the mTEC lineage are temporally separable events that are controlled by distinct mechanisms. We find that whereas mature thymocytes are not essential for Aire(+) mTEC development, use of an inducible ZAP70 transgenic mouse line--in which positive selection can be temporally controlled--demonstrates that the emergence of involucrin(+) mTECs critically depends upon the presence of mature single positive thymocytes. Finally, although initial formation of Aire(+) mTECs depends upon RANK signaling, continued mTEC development to the involucrin(+) stage maps to activation of the LTα-LTβR axis by mature thymocytes. Collectively, our results reveal further complexity in the mechanisms regulating thymus medulla development and highlight the role of distinct TNFRs in initial and terminal differentiation stages in mTECs.
Collapse
Affiliation(s)
- Andrea J. White
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| | - Kyoko Nakamura
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| | - William E. Jenkinson
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| | - Manoj Saini
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| | - Charles Sinclair
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Benedict Seddon
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Parth Narendran
- School of Clinical and Experimental Medicine, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine University, Dusseldorf, Germany
| | - Takeshi Nitta
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima, Japan
| | - Yousuke Takahama
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima, Japan
| | - Jorge H. Caamano
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| | - Peter J. L. Lane
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| | - Eric J. Jenkinson
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| | - Graham Anderson
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham
| |
Collapse
|
48
|
Nishikawa Y, Hirota F, Yano M, Kitajima H, Miyazaki JI, Kawamoto H, Mouri Y, Matsumoto M. Biphasic Aire expression in early embryos and in medullary thymic epithelial cells before end-stage terminal differentiation. J Exp Med 2010; 207:963-71. [PMID: 20404099 PMCID: PMC2867289 DOI: 10.1084/jem.20092144] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 03/03/2010] [Indexed: 12/26/2022] Open
Abstract
The roles of autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) in the organization of the thymic microenvironment for establishing self-tolerance are enigmatic. We sought to monitor the production and maintenance of Aire-expressing mTECs by a fate-mapping strategy in which bacterial artificial chromosome transgenic (Tg) mice expressing Cre recombinase under the control of the Aire regulatory element were crossed with a GFP reporter strain. We found that, in addition to its well recognized expression within mature mTECs, Aire was expressed in the early embryo before emergence of the three germ cell layers. This observation may help to explain the development of ectodermal dystrophy often seen in patients with AIRE deficiency. With the use of one Tg line in which Cre recombinase expression was confined to mTECs, we found that Aire(+)CD80(high) mTECs further progressed to an Aire(-)CD80(intermediate) stage, suggesting that Aire expression is not constitutive from after its induction until cell death but instead is down-regulated at the beginning of terminal differentiation. We also demonstrated that many mTECs of Aire-expressing lineage are in close contact with thymic dendritic cells. This close proximity may contribute to transfer of tissue-restricted self-antigens expressed by mTECs to professional antigen-presenting cells.
Collapse
Affiliation(s)
- Yumiko Nishikawa
- Division of Molecular Immunology, Institute for Enzyme Research, University of Tokushima, Tokushima 770-8503, Japan
| | - Fumiko Hirota
- Division of Molecular Immunology, Institute for Enzyme Research, University of Tokushima, Tokushima 770-8503, Japan
| | - Masashi Yano
- Division of Molecular Immunology, Institute for Enzyme Research, University of Tokushima, Tokushima 770-8503, Japan
| | - Hiroyuki Kitajima
- Division of Human Stem Cell Technology, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Jun-ichi Miyazaki
- Division of Stem Cell Regulation Research (G6), Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Hiroshi Kawamoto
- Laboratory for Lymphocyte Development, RIKEN Research Center for Allergy and Immunology, Kanagawa 230-0045, Japan
| | - Yasuhiro Mouri
- Division of Molecular Immunology, Institute for Enzyme Research, University of Tokushima, Tokushima 770-8503, Japan
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, University of Tokushima, Tokushima 770-8503, Japan
| |
Collapse
|
49
|
Osada M, Jardine L, Misir R, Andl T, Millar SE, Pezzano M. DKK1 mediated inhibition of Wnt signaling in postnatal mice leads to loss of TEC progenitors and thymic degeneration. PLoS One 2010; 5:e9062. [PMID: 20161711 PMCID: PMC2817005 DOI: 10.1371/journal.pone.0009062] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 01/07/2010] [Indexed: 11/24/2022] Open
Abstract
Background Thymic epithelial cell (TEC) microenvironments are essential for the
recruitment of T cell precursors from the bone marrow, as well as the
subsequent expansion and selection of thymocytes resulting in a mature
self-tolerant T cell repertoire. The molecular mechanisms, which control
both the initial development and subsequent maintenance of these critical
microenvironments, are poorly defined. Wnt signaling has been shown to be
important to the development of several epithelial tissues and organs.
Regulation of Wnt signaling has also been shown to impact both early
thymocyte and thymic epithelial development. However, early blocks in thymic
organogenesis or death of the mice have prevented analysis of a role of
canonical Wnt signaling in the maintenance of TECs in the postnatal
thymus. Methodology/Principal Findings Here we demonstrate that tetracycline-regulated expression of the canonical
Wnt inhibitor DKK1 in TECs localized in both the cortex and medulla of adult
mice, results in rapid thymic degeneration characterized by a loss of
ΔNP63+ Foxn1+ and
Aire+ TECs, loss of K5K8DP TECs thought to represent
or contain an immature TEC progenitor, decreased TEC proliferation and the
development of cystic structures, similar to an aged thymus. Removal of DKK1
from DKK1-involuted mice results in full recovery, suggesting that canonical
Wnt signaling is required for the differentiation or proliferation of TEC
populations needed for maintenance of properly organized adult thymic
epithelial microenvironments. Conclusions/Significance Taken together, the results of this study demonstrate that canonical Wnt
signaling within TECs is required for the maintenance of epithelial
microenvironments in the postnatal thymus, possibly through effects on TEC
progenitor/stem cell populations. Downstream targets of Wnt signaling, which
are responsible for maintenance of these TEC progenitors may provide useful
targets for therapies aimed at counteracting age associated thymic
involution or the premature thymic degeneration associated with cancer
therapy and bone marrow transplants.
Collapse
Affiliation(s)
- Masako Osada
- Department of Biology, The City College of New York, New York, New York,
United States of America
| | - Logan Jardine
- Department of Biology, The City College of New York, New York, New York,
United States of America
| | - Ruth Misir
- Department of Biology, The City College of New York, New York, New York,
United States of America
| | - Thomas Andl
- Vanderbilt University Medical Center, Nashville, Tennessee, United States
of America
| | - Sarah E. Millar
- Departments of Dermatology and Cell and Developmental Biology, University
of Pennsylvania, Philadelphia, Pennsylvania, United States of
America
| | - Mark Pezzano
- Department of Biology, The City College of New York, New York, New York,
United States of America
- * E-mail:
| |
Collapse
|
50
|
Milićević Z, Milićević NM, Laan M, Peterson P, Kisand K, Scott HS, Westermann J. Ultrastructure of medullary thymic epithelial cells of autoimmune regulator (Aire)-deficient mice. Immunol Cell Biol 2010; 88:50-6. [PMID: 19721455 DOI: 10.1038/icb.2009.55] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The significance of the autoimmune regulator (Aire) transcription regulator in establishing central tolerance has recently been elucidated in great detail. Still, the role of Aire in medullary thymic epithelial cell (mTEC) physiology is not fully understood. To shed more light on this issue, we studied the ultrastructure of mTECs in Aire-deficient thymus. We show that all types of mTECs show ultrastructural signs of activation and increased intracellular traffic, which suggests that in the absence of Aire their physiology is impaired. Type 6 'large' mTECs are fully developed in Aire-deficient mice and more frequent than in the normal thymus. The frequency of type 5 'undifferentiated' mTECs is also increased. Collectively, our results suggest that the role of Aire in the physiology of mTECs could be more profound and not restricted only to the presentation of self-tissue-restricted antigens and/or apoptosis of end-stage fully mature cell types.
Collapse
Affiliation(s)
- Zivana Milićević
- Faculty of Medicine, Institute of Histology and Embryology, University of Beograd, Beograd, Serbia
| | | | | | | | | | | | | |
Collapse
|