1
|
Miller CN, Waterfield MR, Gardner JM, Anderson MS. Aire in Autoimmunity. Annu Rev Immunol 2024; 42:427-53. [PMID: 38360547 PMCID: PMC11774315 DOI: 10.1146/annurev-immunol-090222-101050] [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] [Indexed: 02/17/2024]
Abstract
The role of the autoimmune regulator (Aire) in central immune tolerance and thymic self-representation was first described more than 20 years ago, but fascinating new insights into its biology continue to emerge, particularly in the era of advanced single-cell genomics. We briefly describe the role of human genetics in the discovery of Aire, as well as insights into its function gained from genotype-phenotype correlations and the spectrum of Aire-associated autoimmunity-including insights from patients with Aire mutations with broad and diverse implications for human health. We then highlight emerging trends in Aire biology, focusing on three topic areas. First, we discuss medullary thymic epithelial diversity and the role of Aire in thymic epithelial development. Second, we highlight recent developments regarding the molecular mechanisms of Aire and its binding partners. Finally, we describe the rapidly evolving biology of the identity and function of extrathymic Aire-expressing cells (eTACs), and a novel eTAC subset called Janus cells, as well as their potential roles in immune homeostasis.
Collapse
Affiliation(s)
- Corey N Miller
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Medicine, University of California, San Francisco, California, USA
| | - Michael R Waterfield
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - James M Gardner
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Surgery, University of California, San Francisco, California, USA
| | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
2
|
Duan A, Liu R, Liu C, Wu F, Su H, Zhou S, Huang M, Tian X, Jia H, Liu Y, Li M, Du H. Mutation of the gene encoding the PHD-type transcription factor SAB23 confers submergence tolerance in rice. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:168-179. [PMID: 37798132 DOI: 10.1093/jxb/erad388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023]
Abstract
Submergence is a major constraint on rice production in South and Southeast Asia. In this study, we determined that a gene of the Sub1A-binding protein family, SAB23, encodes a plant homeodomain (PHD)-type transcription factor that has a novel function of negatively regulating submergence tolerance in rice. The T-DNA insertion mutant sab23 displayed reduced plant height, delayed seed maturation, and lower percentage seed set. Importantly, this mutant also exhibited enhanced submergence tolerance. In addition, CRISPR/Cas9 knock out of SAB23 resulted in a significant reduction in the content of the gibberellin GA4 and a dramatic increase in the content of GA1 in the plants. SAB23 binds to the promoter of CYTOCHROME P450 714B2 (CYP714B2), which encodes a GA13-oxidase that catalyses the conversion of GA53 to GA19. Disruption of SAB23 function led to increased CYP714B2 transcription, and overexpression of CYP714B2 produced phenotypes similar to those of the SAB23-knockout plants. Taken together, our results reveal that SAB23 negatively regulates rice submergence tolerance by modulating CYP714B2 expression, which has significant potential for use in future breeding.
Collapse
Affiliation(s)
- Ao Duan
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
| | - Rui Liu
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
| | - Changchang Liu
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
| | - Fei Wu
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
| | - Hang Su
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
| | - Shuangzhen Zhou
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
| | - Min Huang
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
| | - Xiaohai Tian
- Hubei Collaborative Innovation Center for Grain Crops, Yangzte University, Jingzhou 434025, P.R. China
| | - Haitao Jia
- Hubei Key Laboratory of Food Crop Germplasm and Genetic Improvement, Food Crops Institute, Hubei Academy of Agricultural Sciences, Wuhan, Hubei 430064, P.R. China
| | - Ya Liu
- Maize Research Institute, Beijing Academy of Agriculture & Forestry Sciences, Beijing 100097, P.R. China
| | - Manfei Li
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
| | - Hewei Du
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, P.R. China
- Hubei Collaborative Innovation Center for Grain Crops, Yangzte University, Jingzhou 434025, P.R. China
| |
Collapse
|
3
|
van Laar GG, van Hamburg JP, Tas SW. Extrathymic AIRE-expressing cells: Friends or foes in autoimmunity and cancer? Clin Exp Rheumatol 2022; 21:103141. [PMID: 35840039 DOI: 10.1016/j.autrev.2022.103141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/10/2022] [Indexed: 11/17/2022]
Abstract
Auto-immune regulator (AIRE) is a transcription factor that is mainly known for its crucial role in the thymus. Here, AIRE ensures central tolerance by promoting the expression of peripheral tissue antigens in thymic epithelial cells, which is essential for the negative selection of autoreactive T cells. Intriguingly, AIRE expressing cells have recently been identified in other tissues outside the thymus as well. However, the exact function of these extrathymic AIRE expressing cells (eTACs) remains largely enigmatic. Human eTACs are mainly found in secondary lymphoid tissues under homeostatic conditions, but are also found in pathologies such as the inflamed tissues of patients with autoimmune diseases and in various cancer tissues. eTACs have been demonstrated to express dendritic cell (DC)-like markers, such as MHCII, CD40 and CD127, but also CCR7, IDO and PD-L1. Interestingly, eTACs lack high expression of co-stimulatory molecules, such as CD80 or CD86. In mice, different types of peripheral AIRE expressing cells have been described, including cells with an innate lymphoid cell-like phenotype and antigen presenting cell (APC) function. These findings suggest that eTACs are APCs with the possibility to modulate or inhibit immune responses, which is confirmed by functional murine studies demonstrating the ability of eTACs to induce tolerance in autoreactive T cells. The potential immunomodulatory function of eTACs makes them promising targets to restore tolerance in autoimmunity or improve immunotherapy in cancer settings. Yet, this requires a better understanding of these cells and the molecular mechanisms involved. In this review we aim to summarize the current knowledge and understanding of eTACs, including their putative roles in health and disease.
Collapse
Affiliation(s)
- Gustaaf G van Laar
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Netherlands
| | - Jan Piet van Hamburg
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Netherlands
| | - Sander W Tas
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Netherlands.
| |
Collapse
|
4
|
Oliveira EH, Assis AF, Speck-Hernandez CA, Duarte MJ, Passos GA. Aire Gene Influences the Length of the 3' UTR of mRNAs in Medullary Thymic Epithelial Cells. Front Immunol 2020; 11:1039. [PMID: 32547551 PMCID: PMC7270294 DOI: 10.3389/fimmu.2020.01039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 04/29/2020] [Indexed: 12/15/2022] Open
Abstract
Aire is a transcriptional controller in medullary thymic epithelial cells (mTECs) modulating a set of peripheral tissue antigens (PTAs) and non-PTA mRNAs as well as miRNAs. Even miRNAs exerting posttranscriptional control of mRNAs in mTECs, the composition of miRNA-mRNA networks may differ. Under reduction in Aire expression, networks exhibited greater miRNA diversity controlling mRNAs. Variations in the number of 3'UTR binding sites of Aire-dependent mRNAs may represent a crucial factor that influence the miRNA interaction. To test this hypothesis, we analyzed through bioinformatics the length of 3'UTRs of a large set of Aire-dependent mRNAs. The data were obtained from existing RNA-seq of mTECs of wild type or Aire-knockout (KO) mice. We used computational algorithms as FASTQC, STAR and HTSEQ for sequence alignment and counting reads, DESEQ2 for the differential expression, 3USS for the alternative 3'UTRs and TAPAS for the alternative polyadenylation sites. We identified 152 differentially expressed mRNAs between these samples comprising those that encode PTAs as well as transcription regulators. In Aire KO mTECs, most of these mRNAs featured an increase in the length of their 3'UTRs originating additional miRNA binding sites and new miRNA controllers. Results from the in silico analysis were statistically significant and the predicted miRNA-mRNA interactions were thermodynamically stable. Even with no in vivo or in vitro experiments, they were adequate to show that lack of Aire in mTECs might favor the downregulation of PTA mRNAs and transcription regulators via miRNA control. This could unbalance the overall transcriptional activity in mTECs and thus the self-representation.
Collapse
Affiliation(s)
- Ernna H. Oliveira
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | | | - Cesar A. Speck-Hernandez
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Max Jordan Duarte
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Geraldo A. Passos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, USP, Ribeirão Preto, Brazil
| |
Collapse
|
5
|
Melo‐Lima BL, Poras I, Passos GA, Carosella ED, Donadi EA, Moreau P. The Autoimmune Regulator (Aire) transactivates HLA-G gene expression in thymic epithelial cells. Immunology 2019; 158:121-135. [PMID: 31322727 PMCID: PMC6742766 DOI: 10.1111/imm.13099] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 07/11/2019] [Indexed: 12/12/2022] Open
Abstract
The Autoimmune Regulator (Aire) protein coordinates the negative selection of developing thymocytes by inducing the expression of hundreds of tissue-specific antigens within the thymic medulla, which is also a primary site of the expression of the immune checkpoint HLA-G molecule. Considering the immunomodulatory properties of Aire and HLA-G, and considering that the role of the constitutive thymus expression of HLA-G has not been elucidated, we studied the effect of AIRE cDNA transfection on HLA-G expression in 4D6 thymic cells and in the HLA-G-positive JEG-3 choriocarcinoma cells. Aire promoted the transactivation of HLA-G gene by increasing the overall transcription, inducing the transcription of at least G1 and G2/G4 isoforms, and incrementing the occurrence and distribution of intracellular HLA-G protein solely in 4D6 thymic cells. Luciferase-based assays and chromatin immunoprecipitation experiments performed in 4D6 cells revealed that Aire targeted at least two regions within the 5'-untranslated regulatory region (5'-URR) extending 1·4 kb from the first ATG initiation codon. The interaction occurs independently of three putative Aire-binding sites. These results indicate that the Aire-induced upregulation of HLA-G in thymic cells is likely to act through the interaction of Aire with specific HLA-G 5'-URR DNA-binding factors. Such a multimeric transcriptional complex might operate in the thymus during the process of promiscuous gene expression.
Collapse
Affiliation(s)
- Breno Luiz Melo‐Lima
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
- Division of Clinical ImmunologyDepartment of MedicineRibeirao Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Isabelle Poras
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| | - Geraldo Aleixo Passos
- Molecular Immunogenetics GroupDepartment of GeneticsRibeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Edgardo D. Carosella
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| | - Eduardo Antonio Donadi
- Division of Clinical ImmunologyDepartment of MedicineRibeirao Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Philippe Moreau
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| |
Collapse
|
6
|
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
|
7
|
Gu B, Lambert JP, Cockburn K, Gingras AC, Rossant J. AIRE is a critical spindle-associated protein in embryonic stem cells. eLife 2017; 6:e28131. [PMID: 28742026 PMCID: PMC5560860 DOI: 10.7554/elife.28131] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/17/2017] [Indexed: 12/21/2022] Open
Abstract
Embryonic stem (ES) cells go though embryo-like cell cycles regulated by specialized molecular mechanisms. However, it is not known whether there are ES cell-specific mechanisms regulating mitotic fidelity. Here we showed that Autoimmune Regulator (Aire), a transcription coordinator involved in immune tolerance processes, is a critical spindle-associated protein in mouse ES(mES) cells. BioID analysis showed that AIRE associates with spindle-associated proteins in mES cells. Loss of function analysis revealed that Aire was important for centrosome number regulation and spindle pole integrity specifically in mES cells. We also identified the c-terminal LESLL motif as a critical motif for AIRE's mitotic function. Combined maternal and zygotic knockout further revealed Aire's critical functions for spindle assembly in preimplantation embryos. These results uncovered a previously unappreciated function for Aire and provide new insights into the biology of stem cell proliferation and potential new angles to understand fertility defects in humans carrying Aire mutations.
Collapse
Affiliation(s)
- Bin Gu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | | | - Katie Cockburn
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
8
|
Abramson J, Goldfarb Y. AIRE: From promiscuous molecular partnerships to promiscuous gene expression. Eur J Immunol 2016; 46:22-33. [PMID: 26450177 DOI: 10.1002/eji.201545792] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 08/10/2015] [Accepted: 10/05/2015] [Indexed: 12/11/2022]
Abstract
Autoimmune regulator (AIRE) is a unique transcriptional regulator that induces promiscuous expression of thousands of tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs), a step critical for the induction of immunological self-tolerance. The past 15 years have seen dramatic progress in our understanding of how AIRE induces immunological self-tolerance on a molecular level. This major advancement can be greatly attributed to the identification of a large variety of proteins that physically associate with AIRE, supporting and regulating its transcription-transactivation capacity. These diverse molecular partnerships have been shown to play roles in shuttling AIRE to the nucleus, securing AIRE's interaction with nuclear matrix and chromatin, releasing RNA polymerase-II from its stalled state and potentiating AIRE-mediated gene expression, among others. In this review we discuss the relationship of AIRE with its vast and rather diverse repertoire of partners and highlight how such "promiscuous partnerships" contribute to the phenomenon of "promiscuous gene expression" in the thymus.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Goldfarb
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
9
|
Maslovskaja J, Saare M, Liiv I, Rebane A, Peterson P. Extended HSR/CARD domain mediates AIRE binding to DNA. Biochem Biophys Res Commun 2015; 468:913-20. [PMID: 26607109 DOI: 10.1016/j.bbrc.2015.11.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 11/12/2015] [Indexed: 02/06/2023]
Abstract
Autoimmune regulator (AIRE) activates the transcription of many genes in an unusual promiscuous and stochastic manner. The mechanism by which AIRE binds to the chromatin and DNA is not fully understood, and the regulatory elements that AIRE target genes possess are not delineated. In the current study, we demonstrate that AIRE activates the expression of transiently transfected luciferase reporters that lack defined promoter regions, as well as intron and poly(A) signal sequences. Our protein-DNA interaction experiments with mutated AIRE reveal that the intact homogeneously staining region/caspase recruitment domain (HSR/CARD) and amino acids R113 and K114 are key elements involved in AIRE binding to DNA.
Collapse
Affiliation(s)
- Julia Maslovskaja
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, 50411, Estonia.
| | - Mario Saare
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, 50411, Estonia
| | - Ingrid Liiv
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, 50411, Estonia
| | - Ana Rebane
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, 50411, Estonia
| | - Pärt Peterson
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, 50411, Estonia
| |
Collapse
|
10
|
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: 0.6] [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
|
11
|
Bellacchio E, Palma A, Corrente S, Di Girolamo F, Helen Kemp E, Di Matteo G, Comelli L, Carsetti R, Cascioli S, Cancrini C, Fierabracci A. The possible implication of the S250C variant of the autoimmune regulator protein in a patient with autoimmunity and immunodeficiency: in silico analysis suggests a molecular pathogenic mechanism for the variant. Gene 2014; 549:286-294. [PMID: 25068407 DOI: 10.1016/j.gene.2014.07.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 07/14/2014] [Accepted: 07/24/2014] [Indexed: 12/12/2022]
Abstract
Autoimmunity can develop from an often undetermined interplay of genetic and environmental factors. Rare forms of autoimmune conditions may also result from single gene mutations as for autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, an autosomal recessive disease associated with mutated forms of the autoimmune regulator gene. It was proposed that genetic variability in the autoimmune regulator locus, in particular heterozygous loss-of-function mutations, might favor the development of organ-specific autoimmunity by affecting the presentation of self-antigens in the thymus. Indeed, heterozygous mutations of the autoimmune regulator gene were reported in patients with organ-specific autoimmunity. Also, in primary immunodeficiencies, a breakdown in central/peripheral tolerance frequently produces association with autoimmunity. The causative link may involve a common genetic background and several gene defects have been identified as putative culprits. We report a unique patient, a 14 year old male from Lazio region, affected by common variable immunodeficiency associated with autoimmune manifestations (alopecia, onychodystrophy) and heterozygote for the S250C variant located in the SAND domain of the autoimmune regulator gene protein. To our knowledge this is the first report of the S250C variant in a patient bearing this unusual combination of autoimmunity and immunodeficiency. To obtain insights into the possible molecular effects of the S250C variant, we have carried out an in silico analysis of the SAND domain structure of the autoimmune regulator protein. In particular, homology modeling has allowed us to observe that the cysteine introduced by the S250C variant is surrounded by cationic residues, and by means of molecular dynamics simulations together with pKa calculations, we have shown that these residues remain stably proximal to cysteine-250 lowering its pKa and thus conferring high chemical reactivity to the mutated residue. We propose that the enhanced reactivity of cysteine-250, which is likely to impair the protein function but probably insufficient to produce alone a phenotype as a heterozygous S250C variant due to compensation mechanisms, might become manifest when combined with other genetic/environmental factors. These results can provide the rationale for the patient's unusual phenotype, shedding new light into the pathogenesis of the clinical association of autoimmunity and immunodeficiency.
Collapse
Affiliation(s)
- Emanuele Bellacchio
- Research Laboratories, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Alessia Palma
- Immunology and Pharmacotherapy Area, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Stefania Corrente
- University Department of Paediatrics, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Francesco Di Girolamo
- Department of Laboratory Medicine, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - E Helen Kemp
- Department of Human Metabolism, The Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Gigliola Di Matteo
- Department of Systems Medicine, Tor Vergata University of Rome, Viale Oxford 81, 00133 Rome, Italy
| | - Laura Comelli
- Proteomics Laboratory Istituto di Fisiologia Clinica, CNR Via Moruzzi, 1, 56124 Pisa, Italy
| | - Rita Carsetti
- Immunology and Pharmacotherapy Area, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Simona Cascioli
- Immunology and Pharmacotherapy Area, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Caterina Cancrini
- University Department of Paediatrics, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Alessandra Fierabracci
- Immunology and Pharmacotherapy Area, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy.
| |
Collapse
|
12
|
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: 32] [Impact Index Per Article: 2.9] [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
|
13
|
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
|
14
|
Zumer K, Saksela K, Peterlin BM. The mechanism of tissue-restricted antigen gene expression by AIRE. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:2479-82. [PMID: 23456700 DOI: 10.4049/jimmunol.1203210] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
The autoimmune regulator is a critical transcription factor for generating central tolerance in the thymus. Recent studies have revealed how the autoimmune regulator targets many otherwise tissue-restricted Ag genes to enable negative selection of autoreactive T cells.
Collapse
Affiliation(s)
- Kristina Zumer
- Department of Virology, Haartman Institute, Helsinki University Central Hospital, University of Helsinki, FIN-00014 Helsinki, Finland
| | | | | |
Collapse
|
15
|
Saare M, Rebane A, Rajashekar B, Vilo J, Peterson P. Autoimmune regulator is acetylated by transcription coactivator CBP/p300. Exp Cell Res 2012; 318:1767-78. [PMID: 22659170 DOI: 10.1016/j.yexcr.2012.04.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 04/12/2012] [Accepted: 04/17/2012] [Indexed: 11/17/2022]
Abstract
The Autoimmune Regulator (AIRE) is a regulator of transcription in the thymic medulla, where it controls the expression of a large set of peripheral-tissue specific genes. AIRE interacts with the transcriptional coactivator and acetyltransferase CBP and synergistically cooperates with it in transcriptional activation. Here, we aimed to study a possible role of AIRE acetylation in the modulation of its activity. We found that AIRE is acetylated in tissue culture cells and this acetylation is enhanced by overexpression of CBP and the CBP paralog p300. The acetylated lysines were located within nuclear localization signal and SAND domain. AIRE with mutations that mimicked acetylated K243 and K253 in the SAND domain had reduced transactivation activity and accumulated into fewer and larger nuclear bodies, whereas mutations that mimicked the unacetylated lysines were functionally similar to wild-type AIRE. Analogously to CBP, p300 localized to AIRE-containing nuclear bodies, however, the overexpression of p300 did not enhance the transcriptional activation of AIRE-regulated genes. Further studies showed that overexpression of p300 stabilized the AIRE protein. Interestingly, gene expression profiling revealed that AIRE, with mutations mimicking K243/K253 acetylation in SAND, was able to activate gene expression, although the affected genes were different and the activation level was lower from those regulated by wild-type AIRE. Our results suggest that the AIRE acetylation can influence the selection of AIRE activated genes.
Collapse
Affiliation(s)
- Mario Saare
- Molecular Pathology, Institute of General and Molecular Pathology, University of Tartu, 19th Ravila Str, Tartu, Estonia.
| | | | | | | | | |
Collapse
|
16
|
Žumer K, Low AK, Jiang H, Saksela K, Peterlin BM. Unmodified histone H3K4 and DNA-dependent protein kinase recruit autoimmune regulator to target genes. Mol Cell Biol 2012; 32:1354-62. [PMID: 22310661 PMCID: PMC3318594 DOI: 10.1128/mcb.06359-11] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 01/26/2012] [Indexed: 11/20/2022] Open
Abstract
Autoimmune regulator (AIRE) directs the expression of otherwise tissue-restricted antigens (TRAs) in medullary thymic epithelial cells, allowing their presentation to developing T cells, which leads to central tolerance. We addressed the conundrum of how AIRE is recruited to these otherwise silent genes in cells. Our studies confirmed that interactions between AIRE and the unmodified histone H3K4 (H3K4me0) are important for targeting AIRE to the mouse insulin promoter in chromatin. By replacing its H3K4me0-binding module with one that binds to the methylated H3K4me3, we redirected the mutant AIRE.ING protein to an actively transcribed gene. Nevertheless, the mutant AIRE D297A protein, which could not bind to H3K4me0, still activated the human insulin promoter on an episomal plasmid target. This targeting was due to DNA-dependent protein kinase (DNA-PK). Thus, in cells that lacked the catalytic subunit of DNA-PK (DNA-PKcs), the assembly and activity of AIRE on DNA, whether in chromatin or on episomal plasmids, was abrogated. However, by the heterologous tethering of AIRE to DNA, we could restore its activity on a plasmid target in DNA-PKcs-negative cells. Importantly, mutations in the putative DNA-binding residues in its SAND domain had no effect on the transcriptional effects of AIRE. Thus, AIRE is recruited to TRA genes in chromatin via cooperative interactions with H3K4me0 and DNA-PK.
Collapse
Affiliation(s)
- Kristina Žumer
- Department of Virology, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Medicine, Microbiology and Immunology, Rosalind Russell Medical Research Center, University of California San Francisco, San Francisco, California, USA
| | - Audrey K. Low
- Department of Medicine, Microbiology and Immunology, Rosalind Russell Medical Research Center, University of California San Francisco, San Francisco, California, USA
| | - Huimin Jiang
- Department of Medicine, Microbiology and Immunology, Rosalind Russell Medical Research Center, University of California San Francisco, San Francisco, California, USA
- Complete Genomics Inc., Mountain View, California, USA
| | - Kalle Saksela
- Department of Virology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - B. Matija Peterlin
- Department of Virology, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Medicine, Microbiology and Immunology, Rosalind Russell Medical Research Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
17
|
Lovewell T, Tazi-Ahnini R. Models to explore the molecular function and regulation of AIRE. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2011. [DOI: 10.1016/j.ejmhg.2011.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
18
|
Žumer K, Plemenitaš A, Saksela K, Peterlin BM. Patient mutation in AIRE disrupts P-TEFb binding and target gene transcription. Nucleic Acids Res 2011; 39:7908-19. [PMID: 21724609 PMCID: PMC3185428 DOI: 10.1093/nar/gkr527] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 06/06/2011] [Accepted: 06/08/2011] [Indexed: 01/17/2023] Open
Abstract
Autoimmune regulator (AIRE) is a transcription factor that induces the expression of a large subset of otherwise strictly tissue restricted antigens in medullary thymic epithelial cells, thereby enabling their presentation to developing T cells for negative selection. Mutations in AIRE lead to autoimmune-polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), a rare monogenetic disease. Although it has been reported that AIRE interacts with proteins involved in nuclear transport, DNA-damage response, chromatin remodeling, transcription and pre-mRNA-splicing, the precise mechanism of AIRE-induced tissue restricted antigen expression has remained elusive. In this study, we investigated an APECED patient mutation that causes the loss of the extreme C-terminus of AIRE and found that this mutant protein is transcriptionaly inactive. When tethered heterologously to DNA, this domain could stimulate transcription and splicing by itself. Moreover, the loss of this C-terminus disrupted interactions with the positive transcription elongation factor b (P-TEFb). Via P-TEFb, AIRE increased levels of RNA polymerase II on and enhanced pre-mRNA splicing of heterologous and endogenous target genes. Indeed, the inhibition of CDK9, the kinase subunit of P-TEFb, inhibited AIRE-induced pre-mRNA splicing of these genes. Thus, AIRE requires P-TEFb to activate transcription elongation and co-transcriptional processing of target genes.
Collapse
Affiliation(s)
- Kristina Žumer
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| | | | | | | |
Collapse
|
19
|
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
|
20
|
Abstract
The negative selection of self-reactive thymocytes depends on the expression of tissue-specific antigens by medullary thymic epithelial cells. The autoimmune regulator (Aire) protein plays an important role in turning on these antigens, and the absence of even one Aire-induced tissue-specific antigen in the thymus can lead to autoimmunity in the antigen-expressing target organ. Recently, Aire protein has been detected in peripheral lymphoid organs, suggesting that peripheral Aire plays a complementary role here. In these peripheral sites, Aire was found to regulate the expression of a group of tissue-specific antigens that is distinct from those expressed in the thymus. Furthermore, transgenic antigen expression in extrathymic Aire-expressing cells (eTACs) can mediate deletional tolerance, but the immunological relevance of Aire-dependent, endogenous tissue-specific antigens remains to be determined.
Collapse
Affiliation(s)
- Todd C Metzger
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
21
|
Höckner M, Stefanon K, Schuler D, Fantur R, de Vaufleury A, Dallinger R. Coping with cadmium exposure in various ways: the two helicid snails Helix pomatia and Cantareus aspersus share the metal transcription factor-2, but differ in promoter organization and transcription of their Cd-metallothionein genes. ACTA ACUST UNITED AC 2010; 311:776-87. [PMID: 19691054 DOI: 10.1002/jez.564] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Gastropods are able to withstand fluctuating availabilities of nonessential trace elements such as cadmium by induction of Cd-specific metallothionein isoform (Cd-MT) expression. As in other species, the induction mechanism involves the binding of metal-regulatory transcription factors (MTF-1 or MTF-2) to metal responsive elements (MREs) in the MT promoter regions. Cd-dependent transcription of Cd-MT genes was assessed by quantitative real time PCR in two helicid gastropods, Helix pomatia and Cantareus aspersus, over a period of eight days. The promoter regions of the Cd-MT genes of the two species were sequenced and compared regarding the position of MREs and other relevant potential transcription factor binding sites (TFBs). Cd-MT gene transcription is induced after Cd exposure in Helix pomatia and Cantareus aspersus, showing a transient peak in Helix pomatia, contrasting with a persistent induction rate in Cantareus aspersus. Since the existence of MTF-2 was verified in both species, differing transcription patterns of Cd-MT genes must be due to functional differences in their metal-responsive promoter regions. Both promoters contain a proximal cluster of three MREs overlapping with TFBs for the transcriptional regulator Sp1. In contrast to Cantareus aspersus, however, the Cd-MT gene of Helix pomatia hosts an additional distal MRE overlapping with a Sp1 binding site and a CACCC box. Inhibitory effects of MRE overlapping Sp1 binding sites were observed in other MT genes. We therefore suggest that transient Cd-MT transcription upon Cd(2+) exposure in Helix pomatia may be the result of an inhibitory action of the distal MRE cluster.
Collapse
Affiliation(s)
- M Höckner
- Institute of Zoology, University of Innsbruck, Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
22
|
Org T, Rebane A, Kisand K, Laan M, Haljasorg U, Andreson R, Peterson P. AIRE activated tissue specific genes have histone modifications associated with inactive chromatin. Hum Mol Genet 2009; 18:4699-710. [PMID: 19744957 PMCID: PMC2778368 DOI: 10.1093/hmg/ddp433] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 09/08/2009] [Indexed: 02/06/2023] Open
Abstract
The Autoimmune Regulator (AIRE) protein is expressed in thymic medullary epithelial cells, where it promotes the ectopic expression of tissue-restricted antigens needed for efficient negative selection of developing thymocytes. Mutations in AIRE cause APECED syndrome, which is characterized by a breakdown of self-tolerance. The molecular mechanism by which AIRE increases the expression of a variety of different genes remains unknown. Here, we studied AIRE-regulated genes using whole genome expression analysis and chromatin immunoprecipitation. We show that AIRE preferentially activates genes that are tissue-specific and characterized by low levels of initial expression in stably transfected HEK293 cell model and mouse thymic medullary epithelial cells. In addition, the AIRE-regulated genes lack active chromatin marks, such as histone H3 trimethylation (H3K4me3) and acetylation (AcH3), on their promoters. We also show that during activation by AIRE, the target genes acquire histone H3 modifications associated with transcription and RNA polymerase II. In conclusion, our data show that AIRE is able to promote ectopic gene expression from chromatin associated with histone modifications characteristic to inactive genes.
Collapse
Affiliation(s)
- Tõnis Org
- Department of Molecular Pathology, Institute of General and Molecular Pathology and
| | - Ana Rebane
- Department of Molecular Pathology, Institute of General and Molecular Pathology and
| | - Kai Kisand
- Department of Molecular Pathology, Institute of General and Molecular Pathology and
| | - Martti Laan
- Department of Molecular Pathology, Institute of General and Molecular Pathology and
| | - Uku Haljasorg
- Department of Molecular Pathology, Institute of General and Molecular Pathology and
| | - Reidar Andreson
- Department of Bioinformatics, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Department of Molecular Pathology, Institute of General and Molecular Pathology and
| |
Collapse
|
23
|
Gardner JM, Fletcher AL, Anderson MS, Turley SJ. AIRE in the thymus and beyond. Curr Opin Immunol 2009; 21:582-9. [PMID: 19833494 PMCID: PMC2787634 DOI: 10.1016/j.coi.2009.08.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 08/21/2009] [Accepted: 08/25/2009] [Indexed: 01/09/2023]
Abstract
The maintenance of immunologic self-tolerance requires the coordination of multiple complementary systems. Studies of the Autoimmune Regulator (Aire) gene have revealed that Aire promotes self-tolerance partly by inducing the transcription of a wide array of tissue-specific antigens (TSAs), particularly in the thymus. The importance of Aire is highlighted by the fact that patients and mice defective in Aire expression develop a multi-organ autoimmune syndrome. In this review we discuss recent progress in our understanding of Aire's control of immune tolerance at the cellular and molecular levels, and also address the potential importance of Aire expression both in the thymus and in the peripheral lymphoid organs. The detection of both Aire and TSA expression by cell populations outside of the thymus raises the possibility that such expression may play a relevant role in the maintenance of self-tolerance.
Collapse
Affiliation(s)
- James M Gardner
- Diabetes Center and the Department of Medicine, University of California, San Francisco, CA 94143-0540, USA
| | | | | | | |
Collapse
|
24
|
Peterson P, Org T, Rebane A. Transcriptional regulation by AIRE: molecular mechanisms of central tolerance. Nat Rev Immunol 2008; 8:948-57. [PMID: 19008896 PMCID: PMC2785478 DOI: 10.1038/nri2450] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The negative selection of T cells in the thymus is necessary for the maintenance of self tolerance. Medullary thymic epithelial cells have a key function in this process as they express a large number of tissue-specific self antigens that are presented to developing T cells. Mutations in the autoimmune regulator (AIRE) protein cause a breakdown of central tolerance that is associated with decreased expression of self antigens in the thymus. In this Review, we discuss the role of AIRE in the thymus and recent advances in our understanding of how AIRE might function at the molecular level to regulate gene expression.
Collapse
Affiliation(s)
- Pärt Peterson
- Institute of General and Molecular Pathology, University of Tartu, Tartu 5O411, Estonia.
| | | | | |
Collapse
|
25
|
Ferguson BJ, Cooke A, Peterson P, Rich T. Death in the AIRE. Trends Immunol 2008; 29:306-12. [PMID: 18515183 DOI: 10.1016/j.it.2008.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 03/14/2008] [Accepted: 03/14/2008] [Indexed: 12/17/2022]
Abstract
When thymic epithelia begin to synthesize peripheral tissue antigens such as insulin, we are seeing the result of autoimmune regulator (AIRE) activity and the workings of central tolerance. AIRE is an extraordinary protein that repatterns the transcriptome of medullary thymic epithelia (mTECs) to produce a stroma decorated with peripheral self-peptides. These peptidic arrays are used to purge self-reactive T cells, thereby averting autoimmunity. We now propose that an inherently cytotoxic event such as global chromatin modification paves the way for AIRE action. This injury stimulus might impose temporal restrictions for the T-cell education process and is endured, at least transiently, by the unique cellular environment provided by the medullary thymic epithelia.
Collapse
Affiliation(s)
- Brian J Ferguson
- Department of Pathology, Divisions of Immunology and Cellular Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | | | | | | |
Collapse
|
26
|
Peña PV, Hom RA, Hung T, Lin H, Kuo AJ, Wong RPC, Subach OM, Champagne KS, Zhao R, Verkhusha VV, Li G, Gozani O, Kutateladze TG. Histone H3K4me3 binding is required for the DNA repair and apoptotic activities of ING1 tumor suppressor. J Mol Biol 2008; 380:303-12. [PMID: 18533182 DOI: 10.1016/j.jmb.2008.04.061] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Revised: 04/24/2008] [Accepted: 04/24/2008] [Indexed: 11/18/2022]
Abstract
Inhibitor of growth 1 (ING1) is implicated in oncogenesis, DNA damage repair, and apoptosis. Mutations within the ING1 gene and altered expression levels of ING1 are found in multiple human cancers. Here, we show that both DNA repair and apoptotic activities of ING1 require the interaction of the C-terminal plant homeodomain (PHD) finger with histone H3 trimethylated at Lys4 (H3K4me3). The ING1 PHD finger recognizes methylated H3K4 but not other histone modifications as revealed by the peptide microarrays. The molecular mechanism of the histone recognition is elucidated based on a 2.1 A-resolution crystal structure of the PHD-H3K4me3 complex. The K4me3 occupies a deep hydrophobic pocket formed by the conserved Y212 and W235 residues that make cation-pi contacts with the trimethylammonium group. Both aromatic residues are essential in the H3K4me3 recognition, as substitution of these residues with Ala disrupts the interaction. Unlike the wild-type ING1, the W235A mutant, overexpressed in the stable clones of melanoma cells or in HT1080 cells, was unable to stimulate DNA repair after UV irradiation or promote DNA-damage-induced apoptosis, indicating that H3K4me3 binding is necessary for these biological functions of ING1. Furthermore, N216S, V218I, and G221V mutations, found in human malignancies, impair the ability of ING1 to associate with H3K4me3 or to induce nucleotide repair and cell death, linking the tumorigenic activity of ING1 with epigenetic regulation. Together, our findings reveal the critical role of the H3K4me3 interaction in mediating cellular responses to genotoxic stresses and offer new insight into the molecular mechanism underlying the tumor suppressive activity of ING1.
Collapse
Affiliation(s)
- P V Peña
- Department of Pharmacology, University of Colorado Health Sciences Center, 12801 East 17th Avenue, Aurora, CO 80045-0511, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Su MA, Giang K, Žumer K, Jiang H, Oven I, Rinn JL, DeVoss JJ, Johannes KP, Lu W, Gardner J, Chang A, Bubulya P, Chang HY, Peterlin BM, Anderson MS. Mechanisms of an autoimmunity syndrome in mice caused by a dominant mutation in Aire. J Clin Invest 2008; 118:1712-26. [PMID: 18414681 PMCID: PMC2293336 DOI: 10.1172/jci34523] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Accepted: 02/27/2008] [Indexed: 01/08/2023] Open
Abstract
Homozygous loss-of-function mutations in AIRE cause autoimmune polyglandular syndrome type 1 (APS 1), which manifests in a classic triad of hypoparathyroidism, adrenal insufficiency, and candidiasis. Interestingly, a kindred with a specific G228W AIRE variant presented with an autosomal dominant autoimmune phenotype distinct from APS 1. We utilized a novel G228W-knockin mouse model to show that this variant acted in a dominant-negative manner to cause a unique autoimmunity syndrome. In addition, the expression of a large number of Aire-regulated thymic antigens was partially inhibited in these animals, demonstrating the importance of quantitative changes in thymic antigen expression in determining organ-specific autoimmunity. Furthermore, the dominant-negative effect of the G228W variant was exerted through recruitment of WT Aire away from active sites of transcription in the nucleus of medullary thymic epithelial cells in vivo. Together, these results may demonstrate a mechanism by which autoimmune predisposition to phenotypes distinct from APS 1 can be mediated in a dominant-negative fashion by Aire.
Collapse
Affiliation(s)
- Maureen A. Su
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Karen Giang
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Kristina Žumer
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Huimin Jiang
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Irena Oven
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - John L. Rinn
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Jason J. DeVoss
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Kellsey P.A. Johannes
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Wen Lu
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - James Gardner
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Angela Chang
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Paula Bubulya
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Howard Y. Chang
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - B. Matija Peterlin
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Mark S. Anderson
- Diabetes Center,
Department of Pediatrics, and
Department of Medicine, UCSF, San Francisco, California, USA.
Program in Epithelial Biology, Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.
Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| |
Collapse
|
28
|
Kramnik I. Genetic dissection of host resistance to Mycobacterium tuberculosis: the sst1 locus and the Ipr1 gene. Curr Top Microbiol Immunol 2008; 321:123-48. [PMID: 18727490 DOI: 10.1007/978-3-540-75203-5_6] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Genetic variation of the host significantly contributes to dramatic differences in the outcomes of natural infection with virulent Mycobacterium tuberculosis (MTB) in humans, as well as in experimental animal models. Host resistance to tuberculosis is a complex multifactorial genetic trait in which many genetic polymorphisms contribute to the phenotype, while their individual contributions are influenced by gene-gene and gene-environment interactions. The most epidemiologically significant form of tuberculosis infection in humans is pulmonary tuberculosis. Factors that predispose immunocompetent individuals to this outcome, however, are largely unknown. Using an experimental mouse model of infection with virulent MTB for the genetic analysis of host resistance to this pathogen, we have identified several tuberculosis susceptibility loci in otherwise immunocompetent mice. The sst1 locus has been mapped to mouse chromosome 1 and shown to be especially important for control of pulmonary tuberculosis. Rampant progression of tuberculosis infection in the lungs of the sst1-susceptible mouse was associated with the development of necrotic lung lesions, which was prevented by the sst1-resistant allele. Using a positional cloning approach, we have identified a novel host resistance gene, Ipr1, which is encoded within the sst1 locus and mediates innate immunity to the intracellular bacterial pathogens MTB and Listeria monocytogenes. The sst1 locus and the Ipr1 gene participate in control of intracellular multiplication of virulent MTB and have an effect on the infected macrophages' mechanism of cell death. The Ipr1 is an interferon-inducible nuclear protein that dynamically associates with other nuclear proteins in macrophages primed with interferons or infected with MTB. Several of the Ipr1-interacting proteins are known to participate in regulation of transcription, RNA processing, and apoptosis. Further biochemical analysis of the Ipr1-mediated pathway will help delineate a mechanism of innate immunity that is especially important for control of tuberculosis progression in the lungs.
Collapse
Affiliation(s)
- I Kramnik
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Oven I, Brdicková N, Kohoutek J, Vaupotic T, Narat M, Peterlin BM. AIRE recruits P-TEFb for transcriptional elongation of target genes in medullary thymic epithelial cells. Mol Cell Biol 2007; 27:8815-23. [PMID: 17938200 PMCID: PMC2169392 DOI: 10.1128/mcb.01085-07] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 08/17/2007] [Accepted: 10/02/2007] [Indexed: 01/06/2023] Open
Abstract
AIRE is a transcriptional activator that directs the ectopic expression of many tissue-specific genes in medullary thymic epithelial cells, which plays an important role in the negative selection of autoreactive T cells. However, its mechanism of action remains poorly understood. In this study, we found that AIRE regulates the step of elongation rather than initiation of RNA polymerase II. For these effects, AIRE bound and recruited P-TEFb to target promoters in medullary thymic epithelial cells. In these cells, AIRE activated the ectopic transcription of insulin and salivary protein 1 genes. Indeed, by chromatin immunoprecipitation, we found that RNA polymerase II was already engaged on these promoters but was unable to elongate in the absence of AIRE. Moreover, the genetic inactivation of cyclin T1 from P-TEFb abolished the transcription of AIRE-responsive genes and led to lymphocytic infiltration of lacrimal and salivary glands in the CycT1-/- mouse. Our findings reveal critical steps by which AIRE regulates the transcription of genes that control central tolerance in the thymus.
Collapse
Affiliation(s)
- Irena Oven
- Department of Medicine, Rosalind Russell Medical Research Center, University of California-San Francisco, San Francisco, California 94143-0703, USA
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
In 1997, the autoimmune regulator (AIRE) gene was identified as the locus underlying susceptibility to the polyendocrine autoimmune disease known as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED). In the intervening 10 years, it has become increasingly clear that this rare disorder has provided us with an illuminative window on one of the most fundamental processes of the immune system--the establishment and maintenance of self tolerance.
Collapse
Affiliation(s)
- Diane Mathis
- The Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
31
|
Cheng MH, Shum AK, Anderson MS. What's new in the Aire? Trends Immunol 2007; 28:321-7. [PMID: 17556019 DOI: 10.1016/j.it.2007.05.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 04/17/2007] [Accepted: 05/15/2007] [Indexed: 12/29/2022]
Abstract
Unraveling the mechanisms underlying autoimmune disease remains a difficult challenge. Recent lessons learned from the study of AIRE (autoimmune regulator), the gene responsible for the rare monogenic human syndrome APS-1, highlight the power of genetics to reveal disease pathogenesis. With the discovery of AIRE, central tolerance has re-emerged as a crucial check against autoimmunity. Aire-mediated regulation of diverse self-antigens in the thymus serves as a paradigm for the importance of promiscuous gene expression in the prevention of autoimmune disease. Recent characterization of Aire-targeted antigens continues to bear this out. Here, we review the current progress surrounding the role of Aire in central tolerance from a molecular, genetic and developmental basis.
Collapse
Affiliation(s)
- Mickie H Cheng
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
32
|
DeVoss JJ, Anderson MS. Lessons on immune tolerance from the monogenic disease APS1. Curr Opin Genet Dev 2007; 17:193-200. [PMID: 17466510 DOI: 10.1016/j.gde.2007.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Accepted: 04/13/2007] [Indexed: 12/28/2022]
Abstract
Autoimmunity is a complex disease process that results from a breakdown in the ability of the immune system to discriminate self from non-self. One approach to unraveling how autoimmunity occurs is to study monogenic diseases, for which a single gene defect is responsible. Recent work on the monogenic autoimmune disease 'autoimmune polyglandular syndrome type 1' (APS1) and on the causal gene of this disorder--autoimmune regulator (AIRE)--is providing new lessons on how immune tolerance is maintained.
Collapse
Affiliation(s)
- Jason J DeVoss
- University of California San Francisco Diabetes Center, HSW1102, San Francisco, CA 94143-0540, USA.
| | | |
Collapse
|
33
|
Isaac A, Wilcox KW, Taylor JL. SP100B, a repressor of gene expression preferentially binds to DNA with unmethylated CpGs. J Cell Biochem 2006; 98:1106-22. [PMID: 16775843 DOI: 10.1002/jcb.20841] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
SP100A and SP100B are mammalian nuclear proteins encoded by alternatively-spliced transcripts from the SP100 gene. The N-terminal portion of SP100B (aa 1-476) is identical to SP100A and contains an HP1 interaction domain. The C-terminal portion of SP100B (aa 477-688) contains an HMG2 interaction domain and a SAND domain. The SAND domain is a DNA-binding domain identified in several nuclear proteins involved in transcriptional regulation. We have previously reported that SP100B represses expression of genes present on transfected DNA in a SAND domain-dependent manner. The goal of the present study was to characterize the DNA binding properties of full-length SP100B expressed in mammalian cells. SP100B associated with DNA whereas SP100A did not. The SP100B SAND domain was essential for DNA binding. Deletion of the HP1- or HMG2-binding domain had no effect on DNA binding. SP100B preferentially associated with sequences containing CpG dinucleotides. Our results did not reveal any preference of SP100B for bases flanking CpG dinucleotides. The number of CpGs in a DNA sequence and spacing between CpGs influenced SP100B binding, suggesting that multimers of SP100B bind DNA in a cooperative manner. Binding of SP100B was abrogated by methylation of the cytosine residue within the context of the CpG dinucleotide. We propose that the preference of SP100B for non-methylated CpGs provides a mechanism to target SP100B to foreign DNA, including plasmid DNA or viral DNA genomes, most of which are hypomethylated.
Collapse
Affiliation(s)
- Anne Isaac
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | | |
Collapse
|
34
|
Abstract
Recent elucidation of the role of central tolerance in preventing organ-specific autoimmunity has changed our concepts of self/nonself discrimination. This paradigmatic shift is largely attributable to the discovery of promiscuous expression of tissue-restricted self-antigens (TRAs) by medullary thymic epithelial cells (mTECs). TRA expression in mTECs mirrors virtually all tissues of the body, irrespective of developmental or spatio-temporal expression patterns. This review summarizes current knowledge on the cellular and molecular regulation of TRA expression in mTECs, outlines relevant mechanisms of antigen presentation and modes of tolerance induction, and discusses implications for the pathogenesis of autoimmune diseases and other biological processes such as fertility, pregnancy, puberty, and tumor defense.
Collapse
Affiliation(s)
- Bruno Kyewski
- Division of Developmental Immunology, Tumor Immunology Program, German Cancer Research Center, 69120 Heidelberg, Germany.
| | | |
Collapse
|
35
|
Turunen JA, Wessman M, Forsblom C, Kilpikari R, Parkkonen M, Pöntynen N, Ilmarinen T, Ulmanen I, Peltonen L, Groop PH. Association analysis of the AIRE and insulin genes in Finnish type 1 diabetic patients. Immunogenetics 2006; 58:331-8. [PMID: 16552513 DOI: 10.1007/s00251-006-0088-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Accepted: 01/11/2006] [Indexed: 11/28/2022]
Abstract
Mutations in the autoimmune regulator (AIRE) gene cause a recessive Mendelian disorder autoimmune polyendocrinopathy syndrome type 1 (APS-1 or autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy). APS-1 patients develop multiorgan autoimmune diseases including type 1 diabetes (prevalence 12%). The AIRE protein controls the central tolerance induction in the thymus by regulating the expression levels of tissue-specific peripheral antigens, such as insulin. We hypothesized that the insulin gene (INS) polymorphisms together with the AIRE variations may predispose individuals to diabetes. The role of the AIRE gene was tested both independently and on the condition of the INS risk genotype in the Finnish type 1 diabetes sample. A total of 733 type 1 diabetic cases and 735 age- and sex-matched healthy controls were used in the analysis. Five common single nucleotide polymorphisms (SNPs) in the AIRE gene were selected from the public database (dbSNP). The -23HphI polymorphism was used as a surrogate marker for the INS gene promoter repeat. The five genotyped SNPs in the AIRE gene showed no evidence of association with type 1 diabetes. As expected, the INS gene polymorphism -23HphI was significantly associated with susceptibility to type 1 diabetes (P=6.8 x 10(-12), chi(2) test). When the subclass of patients carrying the homozygote genotype of the INS gene was used in the analysis, the AIRE polymorphisms showed no association with the disease. In conclusion, the AIRE gene does not seem to contribute to disease susceptibility in Finnish type 1 diabetic patients, whereas the insulin gene represents a notable risk factor for disease in this population.
Collapse
Affiliation(s)
- Joni A Turunen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki (C318b), University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ilmarinen T, Melén K, Kangas H, Julkunen I, Ulmanen I, Eskelin P. The monopartite nuclear localization signal of autoimmune regulator mediates its nuclear import and interaction with multiple importin alpha molecules. FEBS J 2006; 273:315-24. [PMID: 16403019 DOI: 10.1111/j.1742-4658.2005.05065.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autoimmune regulator (AIRE) is a transcriptional regulator involved in establishing immunological self-tolerance. Mutations in the AIRE gene lead to the development of the autosomal, recessively inherited, organ-specific autoimmune disease, autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED). The AIRE protein is mainly localized in the cell nucleus where it is associated with nuclear bodies. The N-terminal part of AIRE has been previously shown to mediate nuclear localization of the protein. However, the functional nuclear localization signal (NLS) and nuclear import mechanisms of AIRE have not been identified. We show that, although the amino-acid sequence of AIRE contains a potential bipartite NLS consisting of amino acids 110-114 and 131-133, only the latter part constitutes a functional NLS. Furthermore, we show by in vitro binding assays that AIRE interacts with multiple members of the nuclear transport receptor importin alpha family, mainly alpha1, alpha3, and alpha5, and that these interactions depend on the intactness of the Arg-Lys-rich NLS of AIRE. In addition, we found that AIRE binds to the 'minor' NLS-binding site of importin alpha3 and alpha5 proteins consisting of the C-terminal armadillo repeats 7-9. Our findings strongly suggest that the nuclear import of AIRE is mediated by the classical importin alpha/beta pathway through binding to several importin alpha family members.
Collapse
Affiliation(s)
- Tanja Ilmarinen
- Department of Molecular Medicine, National Public Health Institute, Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
37
|
Ilmarinen T, Eskelin P, Halonen M, Rüppell T, Kilpikari R, Torres GD, Kangas H, Ulmanen I. Functional analysis of SAND mutations in AIRE supports dominant inheritance of the G228W mutation. Hum Mutat 2005; 26:322-31. [PMID: 16114041 DOI: 10.1002/humu.20224] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED) is a rare disorder caused by mutations in the autoimmune regulator gene (AIRE) and characterized by a variable combination of organ-specific autoimmune diseases. Studies on AIRE-deficient mice suggest that AIRE is an important factor in the establishment and maintenance of self-tolerance. The AIRE protein contains several structural domains often found in transcriptional regulators and functions as a transcriptional transactivator in vitro. To date, more than 50 patient mutations have been identified in the coding region of the AIRE gene. So far, APECED has been reported to be inherited in an autosomal recessive manner. However, in contrast to all other AIRE mutations, a novel mutation c.682T>G (p.G228W) in the DNA-binding and/or multimerization domain SAND was recently described to be inherited in a dominant fashion. We analyzed the effects of mutant AIRE proteins containing the patient mutations c.682T>G (p.G228W) and c.755C>T (p.P252L) located in the SAND domain on the properties of the wild-type AIRE in a heterozygous situation in vitro. In addition to the patient mutations, we analyzed the effects of a double mutation [c.727A>G;c.728A>C;c.739C>G;c740G>C] (p.K243A;R247A) of positively charged amino acids in the SAND domain. Of the mutants studied, only c.682T>G (p.G228W) mutant changed the subcellular localization and in addition severely disrupted the transactivating capacity of the wild-type AIRE. Our results indicate that the c.682T>G (p.G228W) mutant AIRE protein acts with a dominant negative effect by binding to the wild-type AIRE, thus preventing the protein from forming the complexes needed for transactivation.
Collapse
Affiliation(s)
- Tanja Ilmarinen
- Department of Molecular Medicine, National Public Health Institute, Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Pereira LE, Bostik P, Ansari AA. The development of mouse APECED models provides new insight into the role of AIRE in immune regulation. Clin Dev Immunol 2005; 12:211-6. [PMID: 16295527 PMCID: PMC2275420 DOI: 10.1080/17402520500212589] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy is a rare recessive autoimmune disorder caused by a defect in a single gene called AIRE (autoimmune regulator). Characteristics of this disease include a variable combination of autoimmune endocrine tissue destruction, mucocutaneous candidiasis and ectodermal dystrophies. The development of Aire-knockout mice has provided an invaluable model for the study of this disease. The aim of this review is to briefly highlight the strides made in APECED research using these transgenic murine models, with a focus on known roles of Aire in autoimmunity. The findings thus far are compelling and prompt additional areas of study which are discussed.
Collapse
Affiliation(s)
- Lara E Pereira
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | | | | |
Collapse
|