1
|
Giraud M, Peterson P. The Autoimmune Regulator (AIRE) Gene, The Master Activator of Self-Antigen Expression in the Thymus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:199-221. [PMID: 40067588 DOI: 10.1007/978-3-031-77921-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
It has been more than 20 years since the AIRE gene was discovered. The mutations in the AIRE gene cause a rare and life-threatening autoimmune disease with severe manifestations against a variety of organs. Since the identification of the AIRE gene in 1997, more than two decades of investigations have revealed key insights into the role of AIRE and its mode of action. These studies have shown that AIRE uniquely induces the expression of thousands of tissue-restricted self-antigens in the thymus. These self-antigens are presented to developing T cells, resulting in the deletion of the self-reactive T cells and the generation of regulatory T cells. Thus, AIRE is a master guardian in establishing and maintaining central immune tolerance.
Collapse
Affiliation(s)
- Matthieu Giraud
- INSERM, Nantes Université, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France.
| | - Pärt Peterson
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.
| |
Collapse
|
2
|
Bez P, Ceraudo M, Vianello F, Rattazzi M, Scarpa R. Where AIRE we now? Where AIRE we going? Curr Opin Allergy Clin Immunol 2024; 24:448-456. [PMID: 39440452 PMCID: PMC11537476 DOI: 10.1097/aci.0000000000001041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
PURPOSE OF REVIEW The purpose of the review is to describe the most recent advancement in understanding of the pivotal role of autoimmune regulator ( AIRE ) gene expression in central and peripheral tolerance, and the implications of its impairment in the genetic and pathogenesis of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) manifestations with insight into possible treatment options. RECENT FINDINGS AIRE gene expression has an important role of central and peripheral tolerance. Different AIRE gene mutations cause APECED, whereas polymorphisms and some variants may be implicated in development of other more frequently autoimmune diseases. Impaired negative T cell selection, reduction of T regulatory function, altered germinal center response, activated B cells and production of autoantibodies explain the development of autoimmunity in APECED. Recent data suggest that an excessive interferon-γ response may be the primer driver of the associated organ damage. Therefore, Janus kinase (JAK)-inhibitors may be promising therapies for treatment of broad spectrum of manifestations. SUMMARY AIRE has a pivotal role in immune tolerance. Disruption of this delicate equilibrium results in complex immune perturbation, ranging from severe autoimmunity, like APECED, to more common organ-specific disorders. Therefore, a deeper understanding of the correlation between AIRE function and clinical phenotype is warranted given the potential translational implication in clinical practice.
Collapse
Affiliation(s)
- Patrick Bez
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Treviso
| | - Martina Ceraudo
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Treviso
- Deparment of Medicine (DIMED)
| | - Fabrizio Vianello
- Hematology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Marcello Rattazzi
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Treviso
- Deparment of Medicine (DIMED)
| | - Riccardo Scarpa
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Treviso
- Deparment of Medicine (DIMED)
| |
Collapse
|
3
|
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
|
4
|
Shirafkan F, Hensel L, Rattay K. Immune tolerance and the prevention of autoimmune diseases essentially depend on thymic tissue homeostasis. Front Immunol 2024; 15:1339714. [PMID: 38571951 PMCID: PMC10987875 DOI: 10.3389/fimmu.2024.1339714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
The intricate balance of immune reactions towards invading pathogens and immune tolerance towards self is pivotal in preventing autoimmune diseases, with the thymus playing a central role in establishing and maintaining this equilibrium. The induction of central immune tolerance in the thymus involves the elimination of self-reactive T cells, a mechanism essential for averting autoimmunity. Disruption of the thymic T cell selection mechanisms can lead to the development of autoimmune diseases. In the dynamic microenvironment of the thymus, T cell migration and interactions with thymic stromal cells are critical for the selection processes that ensure self-tolerance. Thymic epithelial cells are particularly significant in this context, presenting self-antigens and inducing the negative selection of autoreactive T cells. Further, the synergistic roles of thymic fibroblasts, B cells, and dendritic cells in antigen presentation, selection and the development of regulatory T cells are pivotal in maintaining immune responses tightly regulated. This review article collates these insights, offering a comprehensive examination of the multifaceted role of thymic tissue homeostasis in the establishment of immune tolerance and its implications in the prevention of autoimmune diseases. Additionally, the developmental pathways of the thymus are explored, highlighting how genetic aberrations can disrupt thymic architecture and function, leading to autoimmune conditions. The impact of infections on immune tolerance is another critical area, with pathogens potentially triggering autoimmunity by altering thymic homeostasis. Overall, this review underscores the integral role of thymic tissue homeostasis in the prevention of autoimmune diseases, discussing insights into potential therapeutic strategies and examining putative avenues for future research on developing thymic-based therapies in treating and preventing autoimmune conditions.
Collapse
|
5
|
Tao W, Ye Z, Wei Y, Wang J, Yang W, Yu G, Xiong J, Jia S. Insm1 regulates mTEC development and immune tolerance. Cell Mol Immunol 2023; 20:1472-1486. [PMID: 37990032 PMCID: PMC10687002 DOI: 10.1038/s41423-023-01102-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023] Open
Abstract
The expression of self-antigens in medullary thymic epithelial cells (mTECs) is essential for the establishment of immune tolerance, but the regulatory network that controls the generation and maintenance of the multitude of cell populations expressing self-antigens is poorly understood. Here, we show that Insm1, a zinc finger protein with known functions in neuroendocrine and neuronal cells, is broadly coexpressed with an autoimmune regulator (Aire) in mTECs. Insm1 expression is undetectable in most mimetic cell populations derived from mTECs but persists in neuroendocrine mimetic cells. Mutation of Insm1 in mice downregulated Aire expression, dysregulated the gene expression program of mTECs, and altered mTEC subpopulations and the expression of tissue-restricted antigens. Consistent with these findings, loss of Insm1 resulted in autoimmune responses in multiple peripheral tissues. We found that Insm1 regulates gene expression in mTECs by binding to chromatin. Interestingly, the majority of the Insm1 binding sites are co-occupied by Aire and enriched in superenhancer regions. Together, our data demonstrate the important role of Insm1 in the regulation of the repertoire of self-antigens needed to establish immune tolerance.
Collapse
Affiliation(s)
- Weihua Tao
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China
- Key Lab of Guangzhou Basic and Translational Research of Pan-Vascular Diseases, Guangzhou, China
| | - Zhihuan Ye
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China
| | - Yiqiu Wei
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China
| | - Jianxue Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weixin Yang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guoxing Yu
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China
| | - Jieyi Xiong
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.
| | - Shiqi Jia
- The First Affiliated Hospital of Jinan University, Guangzhou, China.
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China.
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China.
- Key Lab of Guangzhou Basic and Translational Research of Pan-Vascular Diseases, Guangzhou, China.
| |
Collapse
|
6
|
Yazdanbakhsh K, Provan D, Semple JW. The role of T cells and myeloid-derived suppressor cells in refractory immune thrombocytopenia. Br J Haematol 2023; 203:54-61. [PMID: 37735552 PMCID: PMC11493757 DOI: 10.1111/bjh.19079] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/31/2023] [Indexed: 09/23/2023]
Abstract
Immune thrombocytopenia (ITP) is characterized by a dysregulated immune response against platelets, affecting both their destruction and production. A role for an abnormal T-cell compartment has been established in ITP pathogenesis and treatments that increase platelet counts in patients with ITP have shown improvements in T-cell profiles. On the other hand, patients who were refractory to treatment appear to retain the T-cell abnormalities as before. Myeloid-derived suppressive cells (MDSCs) are also emerging as key contributors to the immune pathology of ITP and response to treatment. In this review, we will discuss how various treatments affect the T-cell and MDSC compartments in ITP. The review will focus on studies that have examined the underlying mechanisms and/or genetic basis responsible for refractoriness to a given treatment and highlight remaining challenges in identifying factors and mechanisms to predict response to treatment.
Collapse
Affiliation(s)
- Karina Yazdanbakhsh
- Laboratory of Complement Biology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, USA
| | - Drew Provan
- Department of Haematology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - John W. Semple
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
- Clinical Immunology and Transfusion Medicine, Office of Medical Services, Region Skane, Lund, Sweden
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
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: 0.5] [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
|
8
|
Matsumoto M, Yoshida H, Tsuneyama K, Oya T, Matsumoto M. Revisiting Aire and tissue-restricted antigens at single-cell resolution. Front Immunol 2023; 14:1176450. [PMID: 37207224 PMCID: PMC10191227 DOI: 10.3389/fimmu.2023.1176450] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
The thymus is a highly specialized organ that plays an indispensable role in the establishment of self-tolerance, a process characterized by the "education" of developing T-cells. To provide competent T-cells tolerant to self-antigens, medullary thymic epithelial cells (mTECs) orchestrate negative selection by ectopically expressing a wide range of genes, including various tissue-restricted antigens (TRAs). Notably, recent advancements in the high-throughput single-cell analysis have revealed remarkable heterogeneity in mTECs, giving us important clues for dissecting the mechanisms underlying TRA expression. We overview how recent single-cell studies have furthered our understanding of mTECs, with a focus on the role of Aire in inducing mTEC heterogeneity to encompass TRAs.
Collapse
Affiliation(s)
- Minoru Matsumoto
- Department of Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima, Japan
- *Correspondence: Minoru Matsumoto,
| | - Hideyuki Yoshida
- YCI Laboratory for Immunological Transcriptomics, RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Oya
- Department of Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima, Japan
| |
Collapse
|
9
|
Michelson DA, Mathis D. Thymic mimetic cells: tolerogenic masqueraders. Trends Immunol 2022; 43:782-791. [PMID: 36008259 PMCID: PMC9509455 DOI: 10.1016/j.it.2022.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Medullary thymic epithelial cells (mTECs) clonally delete or divert autoreactive T cells by ectopically expressing a diverse array of peripheral-tissue antigens (PTAs) within the thymus. Although thymic stromal cells with histological features of extra-thymic cell types, like myocytes or neurons, have been observed by light microscopy since the mid-1800s, most modern work on PTA expression has focused on the transcription factor Aire. Here, we highlight recent work that has refocused attention on such 'misplaced' thymic cells, referred to collectively as thymic mimetic cells. We review the molecular underpinnings of mimetic cells and their roles in establishing T cell tolerance, and we propose that mimetic cells play important roles in autoimmunity. Finally, we suggest future directions for this emerging area.
Collapse
Affiliation(s)
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Santos JC, Dametto M, Masson AP, Faça VM, Bonacin R, Donadi EA, Passos GA. The AIRE G228W mutation disturbs the interaction of AIRE with its partner molecule SIRT1. Front Immunol 2022; 13:948419. [PMID: 36148232 PMCID: PMC9485725 DOI: 10.3389/fimmu.2022.948419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
The autoimmune regulator (AIRE) protein functions as a tetramer, interacting with partner proteins to form the “AIRE complex,” which relieves RNA Pol II stalling in the chromatin of medullary thymic epithelial cells (mTECs). AIRE is the primary mTEC transcriptional controller, promoting the expression of a large set of peripheral tissue antigen genes implicated in the negative selection of self-reactive thymocytes. Under normal conditions, the SIRT1 protein temporarily interacts with AIRE and deacetylates K residues of the AIRE SAND domain. Once the AIRE SAND domain is deacetylated, the binding with SIRT1 is undone, allowing the AIRE complex to proceed downstream with the RNA Pol II to the elongation phase of transcription. Considering that the in silico and in vitro binding of the AIRE SAND domain with SIRT1 provides a powerful model system for studying the dominant SAND G228W mutation mechanism, which causes the autoimmune polyglandular syndrome-1, we integrated computational molecular modeling, docking, dynamics between the whole SAND domain with SIRT1, and surface plasmon resonance using a peptide harboring the 211 to 230 residues of the SAND domain, to compare the structure and energetics of binding/release between AIRE G228 (wild-type) and W228 (mutant) SAND domain to SIRT1. We observed that the G228W mutation in the SAND domain negatively influences the AIRE-SIRT1 interaction. The disturbed interaction might cause a disruption in the binding of the AIRE SAND domain with the SIRT1 catalytic site, impairing the AIRE complex to proceed downstream with RNA Pol II.
Collapse
Affiliation(s)
- Jadson C. Santos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Mariangela Dametto
- Renato Archer Information Technology Center (CTI Brazil), Ministry of Science, Technology and Innovation (MCTI), Campinas, SP, Brazil
| | - Ana Paula Masson
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Vitor M. Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Rodrigo Bonacin
- Renato Archer Information Technology Center (CTI Brazil), Ministry of Science, Technology and Innovation (MCTI), Campinas, SP, Brazil
| | - Eduardo A. Donadi
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Geraldo Aleixo Passos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- *Correspondence: Geraldo Aleixo Passos,
| |
Collapse
|
11
|
Qi Y, Zhang R, Lu Y, Zou X, Yang W. Aire and Fezf2, two regulators in medullary thymic epithelial cells, control autoimmune diseases by regulating TSAs: Partner or complementer? Front Immunol 2022; 13:948259. [PMID: 36110862 PMCID: PMC9468217 DOI: 10.3389/fimmu.2022.948259] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The expression of tissue-specific antigens (TSAs) in medullary thymic epithelial cells (mTECs) is believed to be responsible for the elimination of autoreactive T cells, a critical process in the maintenance of central immune tolerance. The transcription factor autoimmune regulator (Aire) and FEZ family zinc finger 2(Fezf2) play an essential role in driving the expression of TSAs in mTECs, while their deficiency in humans and mice causes a range of autoimmune manifestations, such as type 1 diabetes, Sjögren's syndrome and rheumatoid arthritis. However, because of their regulatory mechanisms, the expression profile of TSAs and their relationship with special autoimmune diseases are still in dispute. In this review, we compare the roles of Aire and Fezf2 in regulating TSAs, with an emphasis on their molecular mechanisms in autoimmune diseases, which provides the foundation for devising improved diagnostic and therapeutic approaches for patients.
Collapse
Affiliation(s)
| | | | | | - Xueyang Zou
- Department of Immunology, College of Basic Medical Sciences, School of Public Health, Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
12
|
Harley ITW, Allison K, Scofield RH. Polygenic autoimmune disease risk alleles impacting B cell tolerance act in concert across shared molecular networks in mouse and in humans. Front Immunol 2022; 13:953439. [PMID: 36090990 PMCID: PMC9450536 DOI: 10.3389/fimmu.2022.953439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Most B cells produced in the bone marrow have some level of autoreactivity. Despite efforts of central tolerance to eliminate these cells, many escape to periphery, where in healthy individuals, they are rendered functionally non-responsive to restimulation through their antigen receptor via a process termed anergy. Broad repertoire autoreactivity may reflect the chances of generating autoreactivity by stochastic use of germline immunoglobulin gene segments or active mechanisms may select autoreactive cells during egress to the naïve peripheral B cell pool. Likewise, it is unclear why in some individuals autoreactive B cell clones become activated and drive pathophysiologic changes in autoimmune diseases. Both of these remain central questions in the study of the immune system(s). In most individuals, autoimmune diseases arise from complex interplay of genetic risk factors and environmental influences. Advances in genome sequencing and increased statistical power from large autoimmune disease cohorts has led to identification of more than 200 autoimmune disease risk loci. It has been observed that autoantibodies are detectable in the serum years to decades prior to the diagnosis of autoimmune disease. Thus, current models hold that genetic defects in the pathways that control autoreactive B cell tolerance set genetic liability thresholds across multiple autoimmune diseases. Despite the fact these seminal concepts were developed in animal (especially murine) models of autoimmune disease, some perceive a disconnect between human risk alleles and those identified in murine models of autoimmune disease. Here, we synthesize the current state of the art in our understanding of human risk alleles in two prototypical autoimmune diseases - systemic lupus erythematosus (SLE) and type 1 diabetes (T1D) along with spontaneous murine disease models. We compare these risk networks to those reported in murine models of these diseases, focusing on pathways relevant to anergy and central tolerance. We highlight some differences between murine and human environmental and genetic factors that may impact autoimmune disease development and expression and may, in turn, explain some of this discrepancy. Finally, we show that there is substantial overlap between the molecular networks that define these disease states across species. Our synthesis and analysis of the current state of the field are consistent with the idea that the same molecular networks are perturbed in murine and human autoimmune disease. Based on these analyses, we anticipate that murine autoimmune disease models will continue to yield novel insights into how best to diagnose, prognose, prevent and treat human autoimmune diseases.
Collapse
Affiliation(s)
- Isaac T. W. Harley
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
- Rheumatology Section, Medicine Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Kristen Allison
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
| | - R. Hal Scofield
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Medical/Research Service, US Department of Veterans Affairs Medical Center, Oklahoma City, OK, United States
| |
Collapse
|
13
|
Michelson DA, Hase K, Kaisho T, Benoist C, Mathis D. Thymic epithelial cells co-opt lineage-defining transcription factors to eliminate autoreactive T cells. Cell 2022; 185:2542-2558.e18. [PMID: 35714609 PMCID: PMC9469465 DOI: 10.1016/j.cell.2022.05.018] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/21/2022] [Accepted: 05/19/2022] [Indexed: 12/13/2022]
Abstract
Medullary thymic epithelial cells (mTECs) ectopically express thousands of peripheral-tissue antigens (PTAs), which drive deletion or phenotypic diversion of self-reactive immature T cells during thymic differentiation. Failure of PTA expression causes multiorgan autoimmunity. By assaying chromatin accessibility in individual mTECs, we uncovered signatures of lineage-defining transcription factors (TFs) for skin, lung, liver, and intestinal cells-including Grhl, FoxA, FoxJ1, Hnf4, Sox8, and SpiB-in distinct mTEC subtypes. Transcriptomic and histologic analyses showed that these subtypes, which we collectively term mimetic cells, expressed PTAs in a biologically logical fashion, mirroring extra-thymic cell types while maintaining mTEC identity. Lineage-defining TFs bound to mimetic-cell open chromatin regions and were required for mimetic cell accumulation, whereas the tolerogenic factor Aire was partially and variably required. Expression of a model antigen in mimetic cells sufficed to induce cognate T cell tolerance. Thus, mTECs co-opt lineage-defining TFs to drive mimetic cell accumulation, PTA expression, and self-tolerance.
Collapse
Affiliation(s)
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Sun IH, Gillis-Buck E, Mackenzie TC, Gardner JM. Thymic and extrathymic Aire-expressing cells in maternal-fetal tolerance. Immunol Rev 2022; 308:93-104. [PMID: 35535447 DOI: 10.1111/imr.13082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 12/16/2022]
Abstract
Healthy pregnancy requires maternal immune tolerance to both fetal and placental tissues which contain a range of self- and non-self-antigens. While many of the components and mechanisms of maternal-fetal tolerance have been investigated in detail and previously and thoroughly reviewed (Erlebacher A. Annu Rev Immunol. 2013;31:387-411), the role of autoimmune regulator (Aire), a critical regulator of central tolerance expressed by medullary thymic epithelial cells (mTECs), has been less explored. Aire is known to facilitate the expression of a range of otherwise tissue-specific antigens (TSAs) in mTECs, and here we highlight recent work showing a role for mTEC-mediated thymic selection in maintaining maternal-fetal tolerance. Recently, however, our group and others have identified additional populations of extrathymic Aire-expressing cells (eTACs) in the secondary lymphoid organs. These hematopoietic antigen-presenting cells possess the ability to induce functional inactivation and/or deletion of cognate T cells, and deletion of maternal eTACs during pregnancy increases T-cell activation in the lymph nodes and lymphocytic infiltration of the uterus, leading to pregnancy complications including intrauterine growth restriction (IUGR) and fetal resorption. In this review, we briefly summarize findings related to essential Aire biology, discuss the known roles of Aire-deficiency related to pregnancy complications and infertility, review the newly discovered role for eTACs in the maintenance of maternal-fetal tolerance-as well as recent work defining eTACs at the single-cell level-and postulate potential mechanisms by which eTACs may regulate this process.
Collapse
Affiliation(s)
- Im-Hong Sun
- Department of Surgery, University of California, San Francisco, California, USA.,Diabetes Center, University of California, San Francisco, California, USA
| | - Eva Gillis-Buck
- Department of Surgery, University of California, San Francisco, California, USA
| | - Tippi C Mackenzie
- Department of Surgery, University of California, San Francisco, California, USA.,Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
| | - James M Gardner
- Department of Surgery, University of California, San Francisco, California, USA.,Diabetes Center, University of California, San Francisco, California, USA
| |
Collapse
|
15
|
Tanaka PP, Oliveira EH, Vieira-Machado MC, Duarte MJ, Assis AF, Bombonato-Prado KF, Passos GA. miR-155 exerts posttranscriptional control of autoimmune regulator (Aire) and tissue-restricted antigen genes in medullary thymic epithelial cells. BMC Genomics 2022; 23:404. [PMID: 35643451 PMCID: PMC9145475 DOI: 10.1186/s12864-022-08631-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 05/13/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The autoimmune regulator (Aire) gene is critical for the appropriate establishment of central immune tolerance. As one of the main controllers of promiscuous gene expression in the thymus, Aire promotes the expression of thousands of downstream tissue-restricted antigen (TRA) genes, cell adhesion genes and transcription factor genes in medullary thymic epithelial cells (mTECs). Despite the increasing knowledge about the role of Aire as an upstream transcriptional controller, little is known about the mechanisms by which this gene could be regulated. RESULTS Here, we assessed the posttranscriptional control of Aire by miRNAs. The in silico miRNA-mRNA interaction analysis predicted thermodynamically stable hybridization between the 3'UTR of Aire mRNA and miR-155, which was confirmed to occur within the cellular milieu through a luciferase reporter assay. This finding enabled us to hypothesize that miR-155 might play a role as an intracellular posttranscriptional regulator of Aire mRNA. To test this hypothesis, we transfected a murine mTEC cell line with a miR-155 mimic in vitro, which reduced the mRNA and protein levels of Aire. Moreover, large-scale transcriptome analysis showed the modulation of 311 downstream mRNAs, which included 58 TRA mRNAs. Moreover, miR-155 mimic-transfected cells exhibited a decrease in their chemotaxis property compared with control thymocytes. CONCLUSION Overall, the results indicate that miR-155 may posttranscriptionally control Aire mRNA, reducing the respective Aire protein levels; consequently, the levels of mRNAs encode tissue-restricted antigens were affected. In addition, miR-155 regulated a crucial process by which mTECs allow thymocytes' migration through chemotaxis.
Collapse
Affiliation(s)
- Pedro Paranhos Tanaka
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Ernna Hérida Oliveira
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Mayara Cristina Vieira-Machado
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Max Jordan Duarte
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Amanda Freire Assis
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Karina Fittipaldi Bombonato-Prado
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Geraldo Aleixo Passos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil.
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
16
|
Yu T, Xie Y, Yuan J, Gao J, Xiao Z, Wu Y, Chen H. The Nutritional Intervention of Resveratrol Can Effectively Alleviate the Intestinal Inflammation Associated With Celiac Disease Induced by Wheat Gluten. Front Immunol 2022; 13:878186. [PMID: 35450077 PMCID: PMC9017684 DOI: 10.3389/fimmu.2022.878186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Aims Wheat gluten is a critical trigger for celiac disease, often causing inflammatory lesions and oxidative stress damage in the intestines of patients. In daily life, it is difficult for celiac disease patients to strictly avoid the dietary intake of gluten, which makes complementary preventive therapy particularly urgent. As such, we investigated the alleviating effects of resveratrol in vivo and in vitro models of celiac disease. Methods We established in vivo and in vitro models of gluten protein-induced celiac disease. The intervention effect of resveratrol was defined well based on relevant indicators of inflammation, immunity and oxidative stress, and its possible involvement in signaling pathways and genes were also identified. Results Resveratrol was effective in reducing intestinal oxidative stress and inflammatory damage induced by wheat gluten in both cell and mouse models for celiac disease. We identified correlations between the genes (Fgf15, Nr0b2, Aire and Ubd) and signaling pathways (PPAR, AMPK and FoxO) in which resveratrol performed critical roles. Conclusions Resveratrol contributed to regulate development of autoimmunity through up-regulation of Aire and Ubd genes and promote nutrient absorption in intestine through down-regulation of Fgf15 and Nr0b2 genes, as well as played a role in regulating complex response system of oxidative stress, inflammatory response and immune response in intestine by activating PPAR, AMPK and FoxO signaling pathways, thus effectively alleviating the intestinal symptoms of celiac disease.
Collapse
Affiliation(s)
- Tian Yu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China.,Sino-German Joint Research Institute, Nanchang University, Nanchang, China.,School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yiting Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China.,Sino-German Joint Research Institute, Nanchang University, Nanchang, China.,School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Juanli Yuan
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China.,School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Jinyan Gao
- School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Zhiwen Xiao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China.,Sino-German Joint Research Institute, Nanchang University, Nanchang, China.,School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yong Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China.,Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China.,Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Lopes N, Boucherit N, Santamaria JC, Provin N, Charaix J, Ferrier P, Giraud M, Irla M. Thymocytes trigger self-antigen-controlling pathways in immature medullary thymic epithelial stages. eLife 2022; 11:69982. [PMID: 35188458 PMCID: PMC8860447 DOI: 10.7554/elife.69982] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 01/14/2022] [Indexed: 12/20/2022] Open
Abstract
Interactions of developing T cells with Aire+ medullary thymic epithelial cells expressing high levels of MHCII molecules (mTEChi) are critical for the induction of central tolerance in the thymus. In turn, thymocytes regulate the cellularity of Aire+ mTEChi. However, it remains unknown whether thymocytes control the precursors of Aire+ mTEChi that are contained in mTEClo cells or other mTEClo subsets that have recently been delineated by single-cell transcriptomic analyses. Here, using three distinct transgenic mouse models, in which antigen presentation between mTECs and CD4+ thymocytes is perturbed, we show by high-throughput RNA-seq that self-reactive CD4+ thymocytes induce key transcriptional regulators in mTEClo and control the composition of mTEClo subsets, including Aire+ mTEChi precursors, post-Aire and tuft-like mTECs. Furthermore, these interactions upregulate the expression of tissue-restricted self-antigens, cytokines, chemokines, and adhesion molecules important for T-cell development. This gene activation program induced in mTEClo is combined with a global increase of the active H3K4me3 histone mark. Finally, we demonstrate that these self-reactive interactions between CD4+ thymocytes and mTECs critically prevent multiorgan autoimmunity. Our genome-wide study thus reveals that self-reactive CD4+ thymocytes control multiple unsuspected facets from immature stages of mTECs, which determines their heterogeneity.
Collapse
Affiliation(s)
- Noella Lopes
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Nicolas Boucherit
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Jérémy C Santamaria
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Nathan Provin
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jonathan Charaix
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Pierre Ferrier
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Matthieu Giraud
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Magali Irla
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| |
Collapse
|
18
|
Shevyrev D, Tereshchenko V, Kozlov V, Sennikov S. Phylogeny, Structure, Functions, and Role of AIRE in the Formation of T-Cell Subsets. Cells 2022; 11:194. [PMID: 35053310 PMCID: PMC8773594 DOI: 10.3390/cells11020194] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/06/2023] Open
Abstract
It is well known that the most important feature of adaptive immunity is the specificity that provides highly precise recognition of the self, altered-self, and non-self. Due to the high specificity of antigen recognition, the adaptive immune system participates in the maintenance of genetic homeostasis, supports multicellularity, and protects an organism from different pathogens at a qualitatively different level than innate immunity. This seemingly simple property is based on millions of years of evolution that led to the formation of diversification mechanisms of antigen-recognizing receptors and later to the emergence of a system of presentation of the self and non-self antigens. The latter could have a crucial significance because the presentation of nearly complete diversity of auto-antigens in the thymus allows for the "calibration" of the forming repertoires of T-cells for the recognition of self, altered-self, and non-self antigens that are presented on the periphery. The central role in this process belongs to promiscuous gene expression by the thymic epithelial cells that express nearly the whole spectrum of proteins encoded in the genome, meanwhile maintaining their cellular identity. This complex mechanism requires strict control that is executed by several transcription factors. One of the most important of them is AIRE. This noncanonical transcription factor not only regulates the processes of differentiation and expression of peripheral tissue-specific antigens in the thymic medullar epithelial cells but also controls intercellular interactions in the thymus. Besides, it participates in an increase in the diversity and transfer of presented antigens and thus influences the formation of repertoires of maturing thymocytes. Due to these complex effects, AIRE is also called a transcriptional regulator. In this review, we briefly described the history of AIRE discovery, its structure, functions, and role in the formation of antigen-recognizing receptor repertoires, along with other transcription factors. We focused on the phylogenetic prerequisites for the development of modern adaptive immunity and emphasized the importance of the antigen presentation system.
Collapse
Affiliation(s)
- Daniil Shevyrev
- Research Institute for Fundamental and Clinical Immunology (RIFCI), 630099 Novosibirsk, Russia; (V.T.); (V.K.); (S.S.)
| | | | | | | |
Collapse
|
19
|
Martinez-Ruíz GU, Morales-Sánchez A, Bhandoola A. Transcriptional and epigenetic regulation in thymic epithelial cells. Immunol Rev 2022; 305:43-58. [PMID: 34750841 PMCID: PMC8766885 DOI: 10.1111/imr.13034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 01/03/2023]
Abstract
The thymus is required for the development of both adaptive and innate-like T cell subsets. There is keen interest in manipulating thymic function for therapeutic purposes in circumstances of autoimmunity, immunodeficiency, and for purposes of immunotherapy. Within the thymus, thymic epithelial cells play essential roles in directing T cell development. Several transcription factors are known to be essential for thymic epithelial cell development and function, and a few transcription factors have been studied in considerable detail. However, the role of many other transcription factors is less well understood. Further, it is likely that roles exist for other transcription factors not yet known to be important in thymic epithelial cells. Recent progress in understanding of thymic epithelial cell heterogeneity has provided some new insight into transcriptional requirements in subtypes of thymic epithelial cells. However, it is unknown whether progenitors of thymic epithelial cells exist in the adult thymus, and consequently, developmental relationships linking putative precursors with differentiated cell types are poorly understood. While we do not presently possess a clear understanding of stage-specific requirements for transcription factors in thymic epithelial cells, new single-cell transcriptomic and epigenomic technologies should enable rapid progress in this field. Here, we review our current knowledge of transcription factors involved in the development, maintenance, and function of thymic epithelial cells, and the mechanisms by which they act.
Collapse
Affiliation(s)
- Gustavo Ulises Martinez-Ruíz
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Research Division, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
- Children’s Hospital of Mexico Federico Gomez, Mexico City, Mexico
| | - Abigail Morales-Sánchez
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Children’s Hospital of Mexico Federico Gomez, Mexico City, Mexico
| | - Avinash Bhandoola
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
20
|
Shen P, Deng X, Chen Z, Ba X, Qin K, Huang Y, Huang Y, Li T, Yan J, Tu S. SIRT1: A Potential Therapeutic Target in Autoimmune Diseases. Front Immunol 2021; 12:779177. [PMID: 34887866 PMCID: PMC8650132 DOI: 10.3389/fimmu.2021.779177] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/08/2021] [Indexed: 12/21/2022] Open
Abstract
The morbidity and mortality of autoimmune diseases (Ads) have been increasing worldwide, and the identification of novel therapeutic strategies for prevention and treatment is urgently needed. Sirtuin 1 (SIRT1), a member of the class III family of nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, has been reported to participate in the progression of several diseases. SIRT1 also regulates inflammation, oxidative stress, mitochondrial function, immune responses, cellular differentiation, proliferation and metabolism, and its altered functions are likely involved in Ads. Several inhibitors and activators have been shown to affect the development of Ads. SIRT1 may represent a novel therapeutic target in these diseases, and small molecules or natural products that modulate the functions of SIRT1 are potential therapeutic agents. In the present review, we summarize current studies of the biological functions of SIRT1 and its role in the pathogenesis and treatment of Ads.
Collapse
Affiliation(s)
- Pan Shen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Deng
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhe Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xin Ba
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yao Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Yan
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Xiao ZP, Lv T, Hou PP, Manaenko A, Liu Y, Jin Y, Gao L, Jia F, Tian Y, Li P, Zhang JH, Hu Q, Zhang X. Sirtuin 5-Mediated Lysine Desuccinylation Protects Mitochondrial Metabolism Following Subarachnoid Hemorrhage in Mice. Stroke 2021; 52:4043-4053. [PMID: 34807744 DOI: 10.1161/strokeaha.121.034850] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Sirt5 (Sirtuin 5) desuccinylates multiple metabolic enzymes and plays an important role in maintaining energy homeostasis. The goal of this study was to determine whether Sirt5-mediated desuccinylation restores the energy metabolism and protects brain against subarachnoid hemorrhage (SAH). METHODS Male C57BL/6 or Sirt5-/- mice were used. The endovascular perforation SAH model was applied. Protein lysine succinylation in the brain cortex was examined using liquid chromatography-tandem mass spectrometry analysis. The brain metabolism was evaluated by measurement of brain pH as well as ATP and reactive oxygen species level. Neuronal cell death and neurobehavioral deficits were assessed 24 hours after SAH. The expression and desuccinylation activity of Sirt5, lysine succinylation of citrate synthase and ATP synthase subunits were investigated by Western blot, immunohistochemistry, and ELISA in SAH mice and patients. Furthermore, the benefits of resveratrol-mediated Sirt5 activation were investigated. RESULTS A total of 211 lysine succinylation sites were differentially expressed on 170 proteins in mice brain after SAH. Thirty-nine percent of these succinylated proteins were localized in mitochondria and they are related to energy metabolism. SAH caused a decrease of Sirt5 expression and succinylated citrate synthase as well as the subunits of ATP synthase, subsequently lowered brain pH, reduced ATP and increased reactive oxygen species production, leading to neuronal cell death, and neurological deficits. Knockdown of Sirt5 aggravated SAH-induced effects, mentioned above. Administration of resveratrol resulted in activation of Sirt5. The activation was accompanied both with restoration of the mitochondrial metabolism and alleviation of early brain injury as well as with desuccinylating citrate synthase and ATP synthase. CONCLUSIONS Protein lysine succinylation is a biochemical hallmark of metabolic crisis after SAH, and disruption of lysine succinylation through activation of Sirt5 might be a promising therapeutic strategy for the treatment of SAH.
Collapse
Affiliation(s)
- Zhi-Peng Xiao
- Department of Neurosurgery (Z.-P.X., T.L., Y.J., F.J., Q.H., X.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Tao Lv
- Department of Neurosurgery (Z.-P.X., T.L., Y.J., F.J., Q.H., X.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Pin-Pin Hou
- Central Laboratory (P.-P.H., L.G., Q.H.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Anatol Manaenko
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, China (A.M.)
| | - Yuandong Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering East China Normal University, China (Y.L., Y.T.)
| | - Yichao Jin
- Department of Neurosurgery (Z.-P.X., T.L., Y.J., F.J., Q.H., X.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Li Gao
- Central Laboratory (P.-P.H., L.G., Q.H.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Feng Jia
- Department of Neurosurgery (Z.-P.X., T.L., Y.J., F.J., Q.H., X.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering East China Normal University, China (Y.L., Y.T.)
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, China (P.L.)
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, CA (J.H.Z.)
| | - Qin Hu
- Department of Neurosurgery (Z.-P.X., T.L., Y.J., F.J., Q.H., X.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China.,Central Laboratory (P.-P.H., L.G., Q.H.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xiaohua Zhang
- Department of Neurosurgery (Z.-P.X., T.L., Y.J., F.J., Q.H., X.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
22
|
Ferré EMN, Schmitt MM, Lionakis MS. Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy. Front Pediatr 2021; 9:723532. [PMID: 34790633 PMCID: PMC8591095 DOI: 10.3389/fped.2021.723532] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/07/2021] [Indexed: 12/12/2022] Open
Abstract
Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), also known as autoimmune polyglandular syndrome type-1 (APS-1), is a rare monogenic autoimmune disease caused by loss-of-function mutations in the autoimmune regulator (AIRE) gene. AIRE deficiency impairs immune tolerance in the thymus and results in the peripheral escape of self-reactive T lymphocytes and the generation of several cytokine- and tissue antigen-targeted autoantibodies. APECED features a classic triad of characteristic clinical manifestations consisting of chronic mucocutaneous candidiasis (CMC), hypoparathyroidism, and primary adrenal insufficiency (Addison's disease). In addition, APECED patients develop several non-endocrine autoimmune manifestations with variable frequencies, whose recognition by pediatricians should facilitate an earlier diagnosis and allow for the prompt implementation of targeted screening, preventive, and therapeutic strategies. This review summarizes our current understanding of the genetic, immunological, clinical, diagnostic, and treatment features of APECED.
Collapse
Affiliation(s)
| | | | - Michail S. Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
23
|
Jansen K, Shikama-Dorn N, Attar M, Maio S, Lopopolo M, Buck D, Holländer GA, Sansom SN. RBFOX splicing factors contribute to a broad but selective recapitulation of peripheral tissue splicing patterns in the thymus. Genome Res 2021; 31:2022-2034. [PMID: 34649931 PMCID: PMC8559713 DOI: 10.1101/gr.275245.121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022]
Abstract
Thymic epithelial cells (TEC) control the selection of a T cell repertoire reactive to pathogens but tolerant of self. This process is known to involve the promiscuous expression of virtually the entire protein-coding gene repertoire, but the extent to which TEC recapitulate peripheral isoforms, and the mechanisms by which they do so, remain largely unknown. We performed the first assembly-based transcriptomic census of transcript structures and splicing factor (SF) expression in mouse medullary TEC (mTEC) and 21 peripheral tissues. Mature mTEC expressed 60.1% of all protein-coding transcripts, more than was detected in any of the peripheral tissues. However, for genes with tissue-restricted expression, mTEC produced fewer isoforms than did the relevant peripheral tissues. Analysis of exon inclusion revealed an absence of brain-specific microexons in mTEC. We did not find unusual numbers of novel transcripts in TEC, and we show that Aire, the facilitator of promiscuous gene expression, promotes the generation of long “classical” transcripts (with 5′ and 3′ UTRs) but has only a limited impact on alternative splicing in mTEC. Comprehensive assessment of SF expression in mTEC identified a small set of nonpromiscuously expressed SF genes, among which we confirmed RBFOX to be present with AIRE in mTEC nuclei. Using a conditional loss-of-function approach, we show that Rbfox2 promotes mTEC development and regulates the alternative splicing of promiscuously expressed genes. These data indicate that TEC recommission a small number of peripheral SFs, including members of the RBFOX family, to generate a broad but selective representation of the peripheral splice isoform repertoire.
Collapse
Affiliation(s)
- Kathrin Jansen
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom.,Department of Paediatrics and the Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Noriko Shikama-Dorn
- The University Children's Hospital of Basel and the Department of Biomedicine, University of Basel, 4056 Basel, Switzerland
| | - Moustafa Attar
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom.,Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Stefano Maio
- Department of Paediatrics and the Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Maria Lopopolo
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - David Buck
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Georg A Holländer
- Department of Paediatrics and the Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom.,The University Children's Hospital of Basel and the Department of Biomedicine, University of Basel, 4056 Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Stephen N Sansom
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
24
|
Ferrua F, Bortolomai I, Fontana E, Di Silvestre D, Rigoni R, Marcovecchio GE, Draghici E, Brambilla F, Castiello MC, Delfanti G, Moshous D, Picard C, Taghon T, Bordon V, Schulz AS, Schuetz C, Giliani S, Soresina A, Gennery AR, Signa S, Dávila Saldaña BJ, Delmonte OM, Notarangelo LD, Roifman CM, Poliani PL, Uva P, Mauri PL, Villa A, Bosticardo M. Thymic Epithelial Cell Alterations and Defective Thymopoiesis Lead to Central and Peripheral Tolerance Perturbation in MHCII Deficiency. Front Immunol 2021; 12:669943. [PMID: 34211466 PMCID: PMC8239840 DOI: 10.3389/fimmu.2021.669943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/07/2021] [Indexed: 11/13/2022] Open
Abstract
Major Histocompatibility Complex (MHC) class II (MHCII) deficiency (MHCII-D), also known as Bare Lymphocyte Syndrome (BLS), is a rare combined immunodeficiency due to mutations in genes regulating expression of MHCII molecules. MHCII deficiency results in impaired cellular and humoral immune responses, leading to severe infections and autoimmunity. Abnormal cross-talk with developing T cells due to the absence of MHCII expression likely leads to defects in thymic epithelial cells (TEC). However, the contribution of TEC alterations to the pathogenesis of this primary immunodeficiency has not been well characterized to date, in particular in regard to immune dysregulation. To this aim, we have performed an in-depth cellular and molecular characterization of TEC in this disease. We observed an overall perturbation of thymic structure and function in both MHCII-/- mice and patients. Transcriptomic and proteomic profiling of murine TEC revealed several alterations. In particular, we demonstrated that impairment of lymphostromal cross-talk in the thymus of MHCII-/- mice affects mTEC maturation and promiscuous gene expression and causes defects of central tolerance. Furthermore, we observed peripheral tolerance impairment, likely due to defective Treg cell generation and/or function and B cell tolerance breakdown. Overall, our findings reveal disease-specific TEC defects resulting in perturbation of central tolerance and limiting the potential benefits of hematopoietic stem cell transplantation in MHCII deficiency.
Collapse
Affiliation(s)
- Francesca Ferrua
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Ileana Bortolomai
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena Fontana
- Human Genome Department, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Dario Di Silvestre
- Department of Biomedical Sciences, Institute for Biomedical Technologies-National Research Council (CNR), Milan, Italy
| | - Rosita Rigoni
- Human Genome Department, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Genni Enza Marcovecchio
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena Draghici
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Brambilla
- Department of Biomedical Sciences, Institute for Biomedical Technologies-National Research Council (CNR), Milan, Italy
| | - Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Gloria Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Despina Moshous
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker Children’s Hospital, AP-HP, Paris, France
- Laboratory “Genome Dynamics in the Immune System”, INSERM UMR1163, Université de Paris, Institut Imagine, Paris, France
| | - Capucine Picard
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker Children’s Hospital, AP-HP, Paris, France
- Centre d’Etude des Déficits Immunitaires, Necker-Enfants Malades Hospital, AP-HP, Paris, France
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, University Paris Descartes Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Victoria Bordon
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Ansgar S. Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Catharina Schuetz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Silvia Giliani
- Cytogenetics and Medical Genetics Unit and “A. Nocivelli” Institute for Molecular Medicine, Spedali Civili Hospital, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Annarosa Soresina
- Unit of Pediatric Immunology, Pediatrics Clinic, University of Brescia, ASST-Spedali Civili Brescia, Brescia, Italy
| | - Andrew R. Gennery
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Pediatric Immunology and HSCT, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Sara Signa
- Department of Pediatric Immunology and HSCT, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
- Autoinflammatory Diseases and Immunodeficiencies Center, IRCCS Istituto G. Gaslini, and Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal and Children's Sciences, University of Genoa, Genoa, Italy
| | - Blachy J. Dávila Saldaña
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, United States
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, United States
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, United States
| | - Chaim M. Roifman
- Division of Immunology & Allergy, Department of Pediatrics, The Hospital for Sick Children, the Canadian Centre for Primary Immunodeficiency and the University of Toronto, Toronto, ON, Canada
| | - Pietro Luigi Poliani
- Department of Molecular and Translational Medicine, Pathology Unit, University of Brescia, Brescia, Italy
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Pula, Cagliari, Italy
| | - Pier Luigi Mauri
- Department of Biomedical Sciences, Institute for Biomedical Technologies-National Research Council (CNR), Milan, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Marita Bosticardo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, United States
| |
Collapse
|
25
|
Zhang Q, Liang Z, Zhang J, Lei T, Dong X, Su H, Chen Y, Zhang Z, Tan L, Zhao Y. Sirt6 Regulates the Development of Medullary Thymic Epithelial Cells and Contributes to the Establishment of Central Immune Tolerance. Front Cell Dev Biol 2021; 9:655552. [PMID: 33869219 PMCID: PMC8044826 DOI: 10.3389/fcell.2021.655552] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
Although some advances have been made in understanding the molecular regulation of mTEC development, the role of epigenetic regulators in the development and maturation of mTEC is poorly understood. Here, using the TEC-specific Sirt6 knockout mice, we found the deacetylase Sirtuin 6 (Sirt6) is essential for the development of functionally competent mTECs. First of all, TEC-specific Sirt6 deletion dramatically reduces the mTEC compartment, which is caused by reduced DNA replication and subsequent impaired proliferation ability of Sirt6-deficient mTECs. Secondly, Sirt6 deficiency specifically accelerates the differentiation of mTECs from CD80–Aire– immature population to CD80+Aire– intermediate mature population by promoting the expression of Spib. Finally, Sirt6 ablation in TECs markedly interferes the proper expression of tissue-restricted antigens (TRAs) and impairs the development of thymocytes and nTreg cells. In addition, TEC conditional knockout of Sirt6 results in severe autoimmune disease manifested by reduced body weight, the infiltration of lymphocytes and the presence of autoantibodies. Collectively, this study reveals that the expression of epigenetic regulator Sirt6 in TECs is crucial for the development and differentiation of mTECs, which highlights the importance of Sirt6 in the establishment of central immune tolerance.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhanfeng Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiayu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Tong Lei
- University of Chinese Academy of Sciences, Beijing, China
| | - Xue Dong
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Huiting Su
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing, China
| | - Yifang Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhaoqi Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Liang Tan
- Center of Organ Transplantation, Second Xiangya Hospital of Central South University, Changsha, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
26
|
Singh M, Srivastava M, Wakode SR, Asthana S. Elucidation of Structural Determinants Delineates the Residues Playing Key Roles in Differential Dynamics and Selective Inhibition of Sirt1-3. J Chem Inf Model 2021; 61:1105-1124. [PMID: 33606530 DOI: 10.1021/acs.jcim.0c01193] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sirt1-3 are the most studied sirtuins, playing a key role in caloric-dependent epigenetic modifications. Since they are localized in distinct cellular compartments and act differently under various pathological conditions, selective inhibition would be a promising strategy to understand their biological function and to discover effective therapeutics. Here, sirtuin's inhibitor Ex527* is used as a probe to speculate the possible root cause of selective inhibition and differential structural dynamics of Sirt1-3. Comparative energetics and mutational studies revealed the criticality of residues I279 and I316 for the Sirt1 selectivity toward Ex527*. Furthermore, essential dynamics and residue network analysis revealed that the side-chain reorientation in residue F190 due to nonconserved residue Y191 played a major role in the formation of an extended selectivity pocket in Sirt2. These changes at the dynamical and residual level, which impact the internal wiring significantly, might help in rationally designing selective inhibitors against Sirt1-3.
Collapse
Affiliation(s)
- Mrityunjay Singh
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana 121001, India.,Delhi Institute of Pharmaceutical Sciences and Research, DPSRU, M.B. Road, Pushp Vihar, Sector 3, New Delhi 110017, India
| | - Mitul Srivastava
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana 121001, India
| | - Sharad R Wakode
- Delhi Institute of Pharmaceutical Sciences and Research, DPSRU, M.B. Road, Pushp Vihar, Sector 3, New Delhi 110017, India
| | - Shailendra Asthana
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana 121001, India
| |
Collapse
|
27
|
Cosway EJ, James KD, Lucas B, Anderson G, White AJ. The thymus medulla and its control of αβT cell development. Semin Immunopathol 2020; 43:15-27. [PMID: 33306154 PMCID: PMC7925449 DOI: 10.1007/s00281-020-00830-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023]
Abstract
αβT cells are an essential component of effective immune responses. The heterogeneity that lies within them includes subsets that express diverse self-MHC-restricted αβT cell receptors, which can be further subdivided into CD4+ helper, CD8+ cytotoxic, and Foxp3+ regulatory T cells. In addition, αβT cells also include invariant natural killer T cells that are very limited in αβT cell receptor repertoire diversity and recognise non-polymorphic CD1d molecules that present lipid antigens. Importantly, all αβT cell sublineages are dependent upon the thymus as a shared site of their development. Ongoing research has examined how the thymus balances the intrathymic production of multiple αβT cell subsets to ensure correct formation and functioning of the peripheral immune system. Experiments in both wild-type and genetically modified mice have been essential in revealing complex cellular and molecular mechanisms that regulate thymus function. In particular, studies have demonstrated the diverse and critical role that the thymus medulla plays in shaping the peripheral T cell pool. In this review, we summarise current knowledge on functional properties of the thymus medulla that enable the thymus to support the production of diverse αβT cell types.
Collapse
Affiliation(s)
- Emilie J Cosway
- Institute of Immunology and Immunotherapy, Floor 4 Institute for Biomedical Research, Medical School, University of Birmingham, Birmingham, B15 2TT, UK
| | - Kieran D James
- Institute of Immunology and Immunotherapy, Floor 4 Institute for Biomedical Research, Medical School, University of Birmingham, Birmingham, B15 2TT, UK
| | - Beth Lucas
- Institute of Immunology and Immunotherapy, Floor 4 Institute for Biomedical Research, Medical School, University of Birmingham, Birmingham, B15 2TT, UK
| | - Graham Anderson
- Institute of Immunology and Immunotherapy, Floor 4 Institute for Biomedical Research, Medical School, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Andrea J White
- Institute of Immunology and Immunotherapy, Floor 4 Institute for Biomedical Research, Medical School, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
28
|
Rasha F, Mims BM, Castro-Piedras I, Barnes BJ, Grisham MB, Rahman RL, Pruitt K. The Versatility of Sirtuin-1 in Endocrinology and Immunology. Front Cell Dev Biol 2020; 8:589016. [PMID: 33330467 PMCID: PMC7717970 DOI: 10.3389/fcell.2020.589016] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Sirtuins belong to the class III family of NAD-dependent histone deacetylases (HDAC) and are involved in diverse physiological processes that range from regulation of metabolism and endocrine function to coordination of immunity and cellular responses to stress. Sirtuin-1 (SIRT1) is the most well-studied family member and has been shown to be critically involved in epigenetics, immunology, and endocrinology. The versatile roles of SIRT1 include regulation of energy sensing metabolic homeostasis, deacetylation of histone and non-histone proteins in numerous tissues, neuro-endocrine regulation via stimulation of hypothalamus-pituitary axes, synthesis and maintenance of reproductive hormones via steroidogenesis, maintenance of innate and adaptive immune system via regulation of T- and B-cell maturation, chronic inflammation and autoimmune diseases. Moreover, SIRT1 is an appealing target in various disease contexts due to the promise of pharmacological and/or natural modulators of SIRT1 activity within the context of endocrine and immune-related disease models. In this review we aim to provide a broad overview on the role of SIRT1 particularly within the context of endocrinology and immunology.
Collapse
Affiliation(s)
- Fahmida Rasha
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Brianyell McDaniel Mims
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Isabel Castro-Piedras
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Betsy J. Barnes
- Laboratory of Autoimmune and Cancer Research, Center for Autoimmune Musculoskeletal and Hematopoietic Disease, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Molecular Medicine and Department of Pediatrics, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, United States
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | | | - Kevin Pruitt
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
29
|
Liu Z, Zhang H, Hu Y, Liu D, Li L, Li C, Wang Q, Huo J, Liu H, Xie N, Huang X, Liu Y, Chen CD, Shi Y, Zhang X. Critical role of histone H3 lysine 27 demethylase Kdm6b in the homeostasis and function of medullary thymic epithelial cells. Cell Death Differ 2020; 27:2843-2855. [PMID: 32346138 PMCID: PMC7493893 DOI: 10.1038/s41418-020-0546-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 01/26/2023] Open
Abstract
Medullary thymic epithelial cells (mTECs) play a central role in the establishment of T cell central immunological tolerance by promiscuously expressing tissue-restricted antigens (TRAs) and presenting them to developing T cells, leading to deletion of T cells responding to self-antigens. However, molecular mechanisms especially epigenetic regulation of mTEC homeostasis and TRA expression remain elusive. Here we show that the H3K27 demethylase Kdm6b is essential to maintain the postnatal thymic medulla by promoting mTEC survival and regulating the expression of TRA genes. Moreover, mice lacking Kdm6b developed pathological autoimmune disorders. Mechanically, Kdm6b exerted its function by reducing repressive H3K27 trimethylation (H3K27me3) at the promoters of anti-apoptotic gene Bcl2 and a set of Aire-dependent TRA genes. Thus, our findings reveal a dual role of Kdm6b in the regulation of mTEC-mediated T cell central tolerance.
Collapse
Affiliation(s)
- Zhi Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
- Nomis Foundation Laboratories for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Haohao Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yiming Hu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Dandan Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lingling Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Cuifeng Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qi Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Junhaohui Huo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hanshao Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ningxia Xie
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xingxu Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Charlie Degui Chen
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yufang Shi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaoren Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
30
|
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
|
31
|
Sirtuin 1: A Dilemma in Transplantation. J Transplant 2020; 2020:9012980. [PMID: 32373350 PMCID: PMC7196964 DOI: 10.1155/2020/9012980] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/15/2020] [Accepted: 03/20/2020] [Indexed: 12/30/2022] Open
Abstract
Sirtuin 1, a member of sirtuin family of histone deacetylase enzymes, has been implicated in a variety of physiologic and pathologic events, including energy metabolism, cell survival, and age-related alterations. In view of the anti-inflammatory properties of sirtuin 1 along with its protective role in ischemia reperfusion injury, it might be considered as contributing to the promotion of transplantation outcome. However, the potential ability of sirtuin 1 to induce malignancies raises some concerns about its overexpression in clinic. Moreover, despite the findings of sirtuin 1 implication in thymic tolerance induction and T regulatory (Treg) cells survival, there is also evidence for its involvement in Treg suppression and in T helper 17 cells differentiation. The identification of sirtuin 1 natural and synthetic activators leads to the proposal of sirtuin 1 as an eligible target for clinical interventions in transplantation. All positive and negative consequences of sirtuin 1 overactivation/overexpression in the allograft should therefore be studied thoroughly. Herein, we summarize previous findings concerning direct and indirect influences of sirtuin 1 manipulation on transplantation.
Collapse
|
32
|
Gan H, Shen T, Chupp DP, Taylor JR, Sanchez HN, Li X, Xu Z, Zan H, Casali P. B cell Sirt1 deacetylates histone and non-histone proteins for epigenetic modulation of AID expression and the antibody response. SCIENCE ADVANCES 2020; 6:eaay2793. [PMID: 32270032 PMCID: PMC7112761 DOI: 10.1126/sciadv.aay2793] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 01/09/2020] [Indexed: 05/27/2023]
Abstract
Activation-induced cytidine deaminase (AID) mediates immunoglobulin class switch DNA recombination (CSR) and somatic hypermutation (SHM), critical processes for maturation of the antibody response. Epigenetic factors, such as histone deacetylases (HDACs), would underpin B cell differentiation stage-specific AID expression. Here, we showed that NAD+-dependent class III HDAC sirtuin 1 (Sirt1) is highly expressed in resting B cells and down-regulated by stimuli inducing AID. B cell Sirt1 down-regulation, deprivation of NAD+ cofactor, or genetic Sirt1 deletion reduced deacetylation of Aicda promoter histones, Dnmt1, and nuclear factor-κB (NF-κB) p65 and increased AID expression. This promoted class-switched and hypermutated T-dependent and T-independent antibody responses or led to generation of autoantibodies. Genetic Sirt1 overexpression, Sirt1 boost by NAD+, or allosteric Sirt1 enhancement by SRT1720 repressed AID expression and CSR/SHM. By deacetylating histone and nonhistone proteins (Dnmt1 and NF-κB p65), Sirt1 transduces metabolic cues into epigenetic changes to play an important B cell-intrinsic role in modulating antibody and autoantibody responses.
Collapse
Affiliation(s)
| | | | - Daniel P. Chupp
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Julia R. Taylor
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Helia N. Sanchez
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Xin Li
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Zhenming Xu
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Hong Zan
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | | |
Collapse
|
33
|
Bella P, Farini A, Banfi S, Parolini D, Tonna N, Meregalli M, Belicchi M, Erratico S, D'Ursi P, Bianco F, Legato M, Ruocco C, Sitzia C, Sangiorgi S, Villa C, D'Antona G, Milanesi L, Nisoli E, Mauri P, Torrente Y. Blockade of IGF2R improves muscle regeneration and ameliorates Duchenne muscular dystrophy. EMBO Mol Med 2020; 12:e11019. [PMID: 31793167 PMCID: PMC6949491 DOI: 10.15252/emmm.201911019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a debilitating fatal X-linked muscle disorder. Recent findings indicate that IGFs play a central role in skeletal muscle regeneration and development. Among IGFs, insulinlike growth factor 2 (IGF2) is a key regulator of cell growth, survival, migration and differentiation. The type 2 IGF receptor (IGF2R) modulates circulating and tissue levels of IGF2 by targeting it to lysosomes for degradation. We found that IGF2R and the store-operated Ca2+ channel CD20 share a common hydrophobic binding motif that stabilizes their association. Silencing CD20 decreased myoblast differentiation, whereas blockade of IGF2R increased proliferation and differentiation in myoblasts via the calmodulin/calcineurin/NFAT pathway. Remarkably, anti-IGF2R induced CD20 phosphorylation, leading to the activation of sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase (SERCA) and removal of intracellular Ca2+ . Interestingly, we found that IGF2R expression was increased in dystrophic skeletal muscle of human DMD patients and mdx mice. Blockade of IGF2R by neutralizing antibodies stimulated muscle regeneration, induced force recovery and normalized capillary architecture in dystrophic mdx mice representing an encouraging starting point for the development of new biological therapies for DMD.
Collapse
Affiliation(s)
- Pamela Bella
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Andrea Farini
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Stefania Banfi
- Hematology Department Fondazione IRCCSDepartment of Oncology and Hemato‐oncologyIstituto Nazionale dei TumoriUniversitá degli Studi di MilanoMilanItaly
| | | | | | - Mirella Meregalli
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Marzia Belicchi
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | | | - Pasqualina D'Ursi
- Institute of Technologies in BiomedicineNational Research Council (ITB‐CNR)MilanItaly
| | | | - Mariella Legato
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Chiara Ruocco
- Department of Medical Biotechnology and Translational MedicineCenter for Study and Research on ObesityMilan UniversityMilanItaly
| | - Clementina Sitzia
- UOC SMEL‐1Scuola di Specializzazione di Patologia Clinica e Biochimica ClinicaUniversità degli Studi di MilanoMilanItaly
| | - Simone Sangiorgi
- Neurosurgery UnitDepartment of SurgeryASST Lariana‐S. Anna HospitalComoItaly
| | - Chiara Villa
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Giuseppe D'Antona
- Department of Public Health, Experimental and Forensic MedicinePavia UniversityPaviaItaly
| | - Luciano Milanesi
- Institute of Technologies in BiomedicineNational Research Council (ITB‐CNR)MilanItaly
| | - Enzo Nisoli
- Department of Medical Biotechnology and Translational MedicineCenter for Study and Research on ObesityMilan UniversityMilanItaly
| | - PierLuigi Mauri
- Institute of Technologies in BiomedicineNational Research Council (ITB‐CNR)MilanItaly
| | - Yvan Torrente
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| |
Collapse
|
34
|
Chadha S, Wang L, Hancock WW, Beier UH. Sirtuin-1 in immunotherapy: A Janus-headed target. J Leukoc Biol 2019; 106:337-343. [PMID: 30605226 PMCID: PMC7477756 DOI: 10.1002/jlb.2ru1118-422r] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/13/2018] [Accepted: 12/16/2018] [Indexed: 12/16/2022] Open
Abstract
Sirtuin-1 (Sirt1), a member of the NAD-dependent sirtuin family of histone/protein deacetylases (HDAC), is an important target for immunotherapy due to its role in deacetylating the transcription factors Foxp3 and thymic retinoid acid receptor related orphan receptor gamma (RORγt). Sirt1 inhibition can increase Foxp3 acetylation and promote the production and functions of Foxp3+ T-regulatory (Treg) cells, whereas the acetylation of RORγt decreases its transcriptional activity DNA binding and decreases the differentiation of proinflammatory Th17 cells. Pharmacologic inhibitors of Sirt1 increase allograft survival and decrease autoimmune colitis and experimental allergic encephalomyelitis. However, in contrast to its role in T cells, Sirt1 has anti-inflammatory effects in myeloid cells, and, context dependent, in Th17 cells. Here, inhibition of Sirt1 can have proinflammatory effects. In addition to effects arising from the central role of Sirt1 in cellular metabolism and NAD-dependent reactions, such proinflammatory effects further complicate the potential of Sirt1 for therapeutic immunosuppression. This review aims to reconcile the opposing literature on pro- and anti-inflammatory effects of Sirt1, provides an overview of the role of Sir1 in the immune system, and discusses the pros and cons associated with inhibiting Sirt1 for control of inflammation and immune responses.
Collapse
Affiliation(s)
- Sakshum Chadha
- Division of Nephrology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, University of Pennsylvania, Philadelphia, PA 19104, USA
- Current address: Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Liqing Wang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Disease, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Disease, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ulf H. Beier
- Division of Nephrology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Constantine GM, Lionakis MS. Lessons from primary immunodeficiencies: Autoimmune regulator and autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. Immunol Rev 2019; 287:103-120. [PMID: 30565240 PMCID: PMC6309421 DOI: 10.1111/imr.12714] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 08/19/2018] [Indexed: 12/12/2022]
Abstract
The discovery of the autoimmune regulator (AIRE) protein and the delineation of its critical contributions in the establishment of central immune tolerance has significantly expanded our understanding of the immunological mechanisms that protect from the development of autoimmune disease. The parallel identification and characterization of patient cohorts with the monogenic disorder autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), which is typically caused by biallelic AIRE mutations, has underscored the critical contribution of AIRE in fungal immune surveillance at mucosal surfaces and in prevention of multiorgan autoimmunity in humans. In this review, we synthesize the current clinical, genetic, molecular and immunological knowledge derived from basic studies in Aire-deficient animals and from APECED patient cohorts. We also outline major advances and research endeavors that show promise for informing improved diagnostic and therapeutic approaches for patients with APECED.
Collapse
Affiliation(s)
- Gregory M Constantine
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
36
|
Toubai T, Tamaki H, Peltier DC, Rossi C, Oravecz-Wilson K, Liu C, Zajac C, Wu J, Sun Y, Fujiwara H, Henig I, Kim S, Lombard DB, Reddy P. Mitochondrial Deacetylase SIRT3 Plays an Important Role in Donor T Cell Responses after Experimental Allogeneic Hematopoietic Transplantation. THE JOURNAL OF IMMUNOLOGY 2018; 201:3443-3455. [PMID: 30389773 DOI: 10.4049/jimmunol.1800148] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/28/2018] [Indexed: 12/22/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) through its graft-versus-tumor (GVT) effects is a curative therapy against many hematological malignancies. However, GVT is linked to harmful graft-versus-host disease (GVHD) after allo-HCT. Both GVT and GVHD require allogeneic T cell responses, which is an energetically costly process that causes oxidative stress. Sirtuin 3 (SIRT3), a mitochondrial histone deacetylase (HDAC), plays an important role in cellular processes through inhibition of reactive oxygen species (ROS). Nonmitochondrial class of HDACs regulate T cell responses, but the role of mitochondrial HDACs, specifically SIRT3, on donor T cell responses after allo-HCT remains unknown. In this study, we report that SIRT3-deficient (SIRT3-/-) donor T cells cause reduced GVHD severity in multiple clinically relevant murine models. The GVHD protective effect of allogeneic SIRT3-/- T cells was associated with a reduction in their activation, reduced CXCR3 expression, and no significant impact on cytokine secretion or cytotoxic functions. Intriguingly, the GVHD protective effect of SIRT3-/- T cells was associated with a reduction in ROS production, which is contrary to the effect of SIRT3 deficiency on ROS production in other cells/tissues and likely a consequence of their deficient activation. Notably, the reduction in GVHD in the gastrointestinal tract was not associated with a substantial reduction in the GVT effect. Collectively, these data reveal that SIRT3 activity promotes allogeneic donor T cell responses and ROS production without altering T cell cytokine or cytolytic functions and identify SIRT3 as a novel target on donor T cells to improve outcomes after allo-HCT.
Collapse
Affiliation(s)
- Tomomi Toubai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - Hiroya Tamaki
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8131, Japan
| | - Daniel C Peltier
- Division of Hematology and Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109
| | - Corinne Rossi
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109.,Department of Pediatric Hematology and Oncology, University Hospital of Heidelberg, Heidelberg 69120, Germany
| | - Katherine Oravecz-Wilson
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, North Bergen, NJ 08903; and
| | - Cynthia Zajac
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - Julia Wu
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - Yaping Sun
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - Hideaki Fujiwara
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - Israel Henig
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - Stephanie Kim
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - David B Lombard
- Department of Pathology and Institute of Gerontology, University of Michigan, Ann Arbor, MI 48109
| | - Pavan Reddy
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109; .,Department of Pediatric Hematology and Oncology, University Hospital of Heidelberg, Heidelberg 69120, Germany
| |
Collapse
|
37
|
Clark-Knowles KV, Dewar-Darch D, Jardine KE, Coulombe J, Daneshmand M, He X, McBurney MW. Modulating SIRT1 activity variously affects thymic lymphoma development in mice. Exp Cell Res 2018; 371:83-91. [PMID: 30059665 DOI: 10.1016/j.yexcr.2018.07.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/25/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022]
Abstract
SIRT1 is a protein deacetylase with a broad range of biological functions, many of which are known to be important in carcinogenesis, however much of the literature regarding the role of SIRT1 in cancer remains conflicting. In this study we assessed the effect of SIRT1 on the initiation and progression of thymic T cell lymphomas. We employed mouse strains in which SIRT1 activity was absent or could be reversibly modulated in conjunction with thymic lymphoma induction using either the N-nitroso-N-methylurea (NMU) carcinogenesis or the nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) transgene. Decreased SIRT1 activity reduced the development of thymic lymphomas in the NMU-treated mice but was permissive for the formation of lung adenomas. Conversely, in the NPM-ALK transgenic mice, decreased SIRT1 activity had a modest promoting effect in the development of thymic lymphomas. The results of the work presented here add to the growing body of evidence that sirt1 is neither an outright oncogene nor a tumor suppressor. These opposing results in two models of the same disease suggest that the influence of sirt1 on carcinogenesis may lie in a role in tumor surveillance.
Collapse
Affiliation(s)
| | - Danielle Dewar-Darch
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada.
| | - Karen E Jardine
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada.
| | - Josée Coulombe
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada.
| | - Manijeh Daneshmand
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada.
| | - Xiaohong He
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada.
| | - Michael W McBurney
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada; Department of Medicine, University of Ottawa, Ottawa, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
38
|
Zhang Y, Thai K, Jin T, Woo M, Gilbert RE. SIRT1 activation attenuates α cell hyperplasia, hyperglucagonaemia and hyperglycaemia in STZ-diabetic mice. Sci Rep 2018; 8:13972. [PMID: 30228292 PMCID: PMC6143559 DOI: 10.1038/s41598-018-32351-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
The NAD+-dependent lysine deacetylase, Sirtuin 1 (SIRT1), plays a central role in metabolic regulation. With type 1 diabetes a disease that is characterised by metabolic dysregulation, we sought to assess the impact of SIRT1 activation in experimental, streptozotocin (STZ)-induced diabetes. CD1 mice with and without STZ-induced diabetes were randomized to receive the SIRT1 activating compound, SRT3025, or vehicle over 20 weeks. Vehicle treated STZ-CD1 mice developed severe hyperglycaemia with near-absent circulating insulin and widespread beta cell loss in association with hyperglucagonaemia and expanded islet alpha cell mass. Without affecting ß-cell mass or circulating insulin, diabetic mice that received SRT3025 had substantially improved glycaemic control with greatly reduced islet α cell mass and lower plasma glucagon concentrations. Consistent with reduced glucagon abundance, the diabetes-associated overexpression of key gluconeogenic enzymes, glucose-6-phosphatase and PEPCK were also lowered by SRT3025. Incubating cultured α cells with SRT3025 diminished their glucagon secretion and proliferative activity in association with a reduction in the α cell associated transcription factor, Aristaless Related Homeobox (Arx). By reducing the paradoxical increase in glucagon, SIRT1 activation may offer a new, α-cell centric approach to the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Yanling Zhang
- St. Michael's Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, M5B 1W8, Canada
| | - Kerri Thai
- St. Michael's Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, M5B 1W8, Canada
| | - Tianru Jin
- Toronto General Hospital Research Institute (TGHRI), Toronto, ON, M5G 2C4, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute (TGHRI), Toronto, ON, M5G 2C4, Canada
| | - Richard E Gilbert
- St. Michael's Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, M5B 1W8, Canada.
| |
Collapse
|
39
|
Hargarten JC, Williamson PR. Epigenetic Regulation of Autophagy: A Path to the Control of Autoimmunity. Front Immunol 2018; 9:1864. [PMID: 30154791 PMCID: PMC6102341 DOI: 10.3389/fimmu.2018.01864] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
Autoimmune diseases are a significant cause of debilitation and mortality globally and are in need of cost-effective therapeutics. Autophagy is a cellular pathway that facilitates immune modulation involved in both pathogen control and autoimmunity. Regulation is multifactorial and includes a number of epigenetic pathways which can involve modification of DNA-binding histones to induce autophagy-related mRNA synthesis or microRNA and decapping-associated mRNA degradation which results in autophagy suppression. Appreciation of epigenetic-based pathways involved in autophagy and autoimmunity may facilitate application of a burgeoning group of epigenetic pharmaceuticals to these important diseases.
Collapse
Affiliation(s)
- Jessica C Hargarten
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Peter R Williamson
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
40
|
Ossart J, Moreau A, Autrusseau E, Ménoret S, Martin JC, Besnard M, Ouisse LH, Tesson L, Flippe L, Kisand K, Peterson P, Hubert FX, Anegon I, Josien R, Guillonneau C. Breakdown of Immune Tolerance in AIRE-Deficient Rats Induces a Severe Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy-like Autoimmune Disease. THE JOURNAL OF IMMUNOLOGY 2018; 201:874-887. [PMID: 29959280 DOI: 10.4049/jimmunol.1701318] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 06/02/2018] [Indexed: 12/31/2022]
Abstract
Autoimmune regulator (AIRE) deficiency in humans induces a life-threatening generalized autoimmune disease called autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), and no curative treatments are available. Several models of AIRE-deficient mice have been generated, and although they have been useful in understanding the role of AIRE in central tolerance, they do not reproduce accurately the APECED symptoms, and thus there is still a need for an animal model displaying APECED-like disease. We assessed, in this study, the potential of the rat as an accurate model for APECED. In this study, we demonstrate that in rat, AIRE is expressed by MHC class II (MCH-II)+ and MHC-II- medullary thymic epithelial cells in thymus and by CD4int conventional dendritic cells in periphery. To our knowledge, we generated the first AIRE-deficient rat model using zinc-finger nucleases and demonstrated that they display several of the key symptoms of APECED disease, including alopecia, skin depigmentation, and nail dystrophy, independently of the genetic background. We observed severe autoimmune lesions in a large spectrum of organs, in particular in the pancreas, and identified several autoantibodies in organs and cytokines such as type I IFNs and IL-17 at levels similar to APECED. Finally, we demonstrated a biased Ab response to IgG1, IgM, and IgA isotypes. Altogether, our data demonstrate that AIRE-deficient rat is a relevant APECED animal model, opening new opportunity to test curative therapeutic treatments.
Collapse
Affiliation(s)
- Jason Ossart
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France
| | - Anne Moreau
- Anatomie et Cytologie Pathologiques, CHU Nantes, 44093 Nantes, France
| | - Elodie Autrusseau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France
| | - Séverine Ménoret
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France.,Transgenesis Rat Immunophenomic Platform, INSERM 1064 and SFR Francois Bonamy, CNRS UMS3556, 44093 Nantes, France
| | - Jérôme C Martin
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France
| | - Marine Besnard
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France
| | - Laure-Hélène Ouisse
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France.,Transgenesis Rat Immunophenomic Platform, INSERM 1064 and SFR Francois Bonamy, CNRS UMS3556, 44093 Nantes, France
| | - Laurent Tesson
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France.,Transgenesis Rat Immunophenomic Platform, INSERM 1064 and SFR Francois Bonamy, CNRS UMS3556, 44093 Nantes, France
| | - Léa Flippe
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France
| | - Kai Kisand
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia; and
| | - Pärt Peterson
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia; and
| | - François-Xavier Hubert
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France.,Transgenesis Rat Immunophenomic Platform, INSERM 1064 and SFR Francois Bonamy, CNRS UMS3556, 44093 Nantes, France
| | - Régis Josien
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France.,Laboratoire d'Immunologie, CHU Nantes, 44093 Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM Université de Nantes, 44093 Nantes, France; .,Institut de Transplantation Urologie Néphrologie, CHU Nantes, 44093 Nantes, France
| |
Collapse
|
41
|
Cowan JE, Baik S, McCarthy NI, Parnell SM, White AJ, Jenkinson WE, Anderson G. Aire controls the recirculation of murine Foxp3 + regulatory T-cells back to the thymus. Eur J Immunol 2018; 48:844-854. [PMID: 29285761 PMCID: PMC6001551 DOI: 10.1002/eji.201747375] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/07/2017] [Accepted: 12/23/2017] [Indexed: 11/07/2022]
Abstract
In the thymus, medullary thymic epithelial cells (mTEC) determine the fate of newly selected CD4+ and CD8+ single positive (SP) thymocytes. For example, mTEC expression of Aire controls intrathymic self-antigen availability for negative selection. Interestingly, alterations in both Foxp3+ Regulatory T-cells (T-Reg) and conventional SP thymocytes in Aire-/- mice suggest additional, yet poorly understood, roles for Aire during intrathymic T-cell development. To examine this, we analysed thymocytes from Aire-/- mice using Rag2GFP and Foxp3 expression, and a recently described CD69/MHCI subset definition of post-selection CD4+ conventional thymocytes. We show that while Aire is dispensable for de novo generation of conventional αβT-cells, it plays a key role in controlling the intrathymic T-Reg pool. Surprisingly, a decline in intrathymic T-Reg in Aire-/- mice maps to a reduction in mature recirculating Rag2GFP- T-Reg that express CCR6 and re-enter the thymus from the periphery. Furthermore, we show mTEC expression of the CCR6 ligand CCL20 is reduced in Aire-/- mice, and that CCR6 is required for T-Reg recirculation back to the thymus. Collectively, our study re-defines requirements for late stage intrathymic αβT-cell development, and demonstrates that Aire controls a CCR6-CCL20 axis that determines the developmental makeup of the intrathymic T-Reg pool.
Collapse
Affiliation(s)
- Jennifer E. Cowan
- Institute of Immunology and ImmunotherapyCollege of Medical and Dental SciencesMedical SchoolUniversity of BirminghamEdgbastonBirminghamUK
| | - Song Baik
- Institute of Immunology and ImmunotherapyCollege of Medical and Dental SciencesMedical SchoolUniversity of BirminghamEdgbastonBirminghamUK
| | - Nicholas I. McCarthy
- Institute of Immunology and ImmunotherapyCollege of Medical and Dental SciencesMedical SchoolUniversity of BirminghamEdgbastonBirminghamUK
| | - Sonia M. Parnell
- Institute of Immunology and ImmunotherapyCollege of Medical and Dental SciencesMedical SchoolUniversity of BirminghamEdgbastonBirminghamUK
| | - Andrea J. White
- Institute of Immunology and ImmunotherapyCollege of Medical and Dental SciencesMedical SchoolUniversity of BirminghamEdgbastonBirminghamUK
| | - William E. Jenkinson
- Institute of Immunology and ImmunotherapyCollege of Medical and Dental SciencesMedical SchoolUniversity of BirminghamEdgbastonBirminghamUK
| | - Graham Anderson
- Institute of Immunology and ImmunotherapyCollege of Medical and Dental SciencesMedical SchoolUniversity of BirminghamEdgbastonBirminghamUK
| |
Collapse
|
42
|
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
|
43
|
The molecular basis of immune regulation in autoimmunity. Clin Sci (Lond) 2018; 132:43-67. [PMID: 29305419 DOI: 10.1042/cs20171154] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 12/11/2022]
Abstract
Autoimmune diseases can be triggered and modulated by various molecular and cellular characteristics. The mechanisms of autoimmunity and the pathogenesis of autoimmune diseases have been investigated for several decades. It is well accepted that autoimmunity is caused by dysregulated/dysfunctional immune susceptible genes and environmental factors. There are multiple physiological mechanisms that regulate and control self-reactivity, but which can also lead to tolerance breakdown when in defect. The majority of autoreactive T or B cells are eliminated during the development of central tolerance by negative selection. Regulatory cells such as Tregs (regulatory T) and MSCs (mesenchymal stem cells), and molecules such as CTLA-4 (cytotoxic T-lymphocyte associated antigen 4) and IL (interleukin) 10 (IL-10), help to eliminate autoreactive cells that escaped to the periphery in order to prevent development of autoimmunity. Knowledge of the molecular basis of immune regulation is needed to further our understanding of the underlying mechanisms of loss of tolerance in autoimmune diseases and pave the way for the development of more effective, specific, and safer therapeutic interventions.
Collapse
|
44
|
Nishijima H, Kajimoto T, Matsuoka Y, Mouri Y, Morimoto J, Matsumoto M, Kawano H, Nishioka Y, Uehara H, Izumi K, Tsuneyama K, Okazaki IM, Okazaki T, Hosomichi K, Shiraki A, Shibutani M, Mitsumori K, Matsumoto M. Paradoxical development of polymyositis-like autoimmunity through augmented expression of autoimmune regulator (AIRE). J Autoimmun 2018; 86:75-92. [PMID: 28931462 DOI: 10.1016/j.jaut.2017.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/13/2017] [Accepted: 09/13/2017] [Indexed: 11/22/2022]
Abstract
Autoimmunity is prevented by the function of the autoimmune regulator [AIRE (Aire in mice)], which promotes the expression of a wide variety of tissue-restricted antigens (TRAs) from medullary thymic epithelial cells (mTECs) and from a subset of peripheral antigen-presenting cells (APCs). We examined the effect of additive expression of human AIRE (huAIRE) in a model of autoimmune diabetes in NOD mice. Unexpectedly, we observed that mice expressing augmented AIRE/Aire developed muscle-specific autoimmunity associated with incomplete maturation of mTECs together with impaired expression of Aire-dependent TRAs. This led to failure of deletion of autoreactive T cells together with dramatically reduced production of regulatory T cells in the thymus. In peripheral APCs, expression of costimulatory molecules was augmented. We suggest that levels of Aire expression need to be tightly controlled for maintenance of immunological tolerance. Our results also highlight the importance of coordinated action between central tolerance and peripheral tolerance under the common control of Aire.
Collapse
Affiliation(s)
- Hitoshi Nishijima
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Tatsuya Kajimoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Yoshiki Matsuoka
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Yasuhiro Mouri
- 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
| | - Minoru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan; Department of Molecular and Environmental Pathology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - 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, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Hisanori Uehara
- Department of Molecular and Environmental Pathology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Keisuke Izumi
- Department of Molecular and Environmental Pathology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Koichi Tsuneyama
- Department of Molecular and Environmental Pathology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Il-Mi Okazaki
- Division of Immune Regulation, Institute for Genome Research, Tokushima University, Tokushima 770-8503, Japan
| | - Taku Okazaki
- Division of Immune Regulation, Institute for Genome Research, Tokushima University, Tokushima 770-8503, Japan
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-0934, Japan
| | - Ayako Shiraki
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Kunitoshi Mitsumori
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan.
| |
Collapse
|
45
|
Passos GA, Speck‐Hernandez CA, Assis AF, Mendes‐da‐Cruz DA. Update on Aire and thymic negative selection. Immunology 2018; 153:10-20. [PMID: 28871661 PMCID: PMC5721245 DOI: 10.1111/imm.12831] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/24/2017] [Accepted: 08/31/2017] [Indexed: 12/17/2022] Open
Abstract
Twenty years ago, the autoimmune regulator (Aire) gene was associated with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, and was cloned and sequenced. Its importance goes beyond its abstract link with human autoimmune disease. Aire identification opened new perspectives to better understand the molecular basis of central tolerance and self-non-self distinction, the main properties of the immune system. Since 1997, a growing number of immunologists and molecular geneticists have made important discoveries about the function of Aire, which is essentially a pleiotropic gene. Aire is one of the functional markers in medullary thymic epithelial cells (mTECs), controlling their differentiation and expression of peripheral tissue antigens (PTAs), mTEC-thymocyte adhesion and the expression of microRNAs, among other functions. With Aire, the immunological tolerance became even more apparent from the molecular genetics point of view. Currently, mTECs represent the most unusual cells because they express almost the entire functional genome but still maintain their identity. Due to the enormous diversity of PTAs, this uncommon gene expression pattern was termed promiscuous gene expression, the interpretation of which is essentially immunological - i.e. it is related to self-representation in the thymus. Therefore, this knowledge is strongly linked to the negative selection of autoreactive thymocytes. In this update, we focus on the most relevant results of Aire as a transcriptional and post-transcriptional controller of PTAs in mTECs, its mechanism of action, and its influence on the negative selection of autoreactive thymocytes as the bases of the induction of central tolerance and prevention of autoimmune diseases.
Collapse
Affiliation(s)
- Geraldo A. Passos
- Molecular Immunogenetics GroupDepartment of GeneticsRibeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoSPBrazil
- Discipline of Genetics and Molecular BiologyDepartment of Morphology, Physiology and Basic PathologySchool of Dentistry of Ribeirão PretoUniversity of São PauloRibeirão PretoSPBrazil
| | - Cesar A. Speck‐Hernandez
- Graduate Programme in Basic and Applied ImmunologyRibeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoSPBrazil
| | - Amanda F. Assis
- Molecular Immunogenetics GroupDepartment of GeneticsRibeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoSPBrazil
| | - Daniella A. Mendes‐da‐Cruz
- Laboratory on Thymus ResearchOswaldo Cruz InstituteOswaldo Cruz FoundationRio de JaneiroRJBrazil
- National Institute of Science and Technology on NeuroimmunomodulationRio de JaneiroRJBrazil
| |
Collapse
|
46
|
Proekt I, Miller CN, Lionakis MS, Anderson MS. Insights into immune tolerance from AIRE deficiency. Curr Opin Immunol 2017; 49:71-78. [PMID: 29065385 PMCID: PMC5705335 DOI: 10.1016/j.coi.2017.10.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/25/2017] [Accepted: 10/03/2017] [Indexed: 01/07/2023]
Abstract
AIRE is a well-established master regulator of central tolerance. It plays an essential role in driving expression of tissue-specific antigens in the thymus and shaping the development of positively selected T-cells. Humans and mice with compromised or absent AIRE function have markedly variable phenotypes that include a range of autoimmune manifestations. Recent evidence suggests that this variability stems from cooperation of autoimmune susceptibilities involving both central and peripheral tolerance checkpoints. Here we discuss the broadening understanding of the factors that influence Aire expression, modify AIRE function, and the impact and intersection of AIRE with peripheral immunity. This rapidly expanding body of knowledge will force a reexamination of the definition and clinical management of APS-1 patients as well as provide a foundation for the development of immunomodulatory strategies targeting central tolerance.
Collapse
Affiliation(s)
- Irina Proekt
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Corey N Miller
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michail S Lionakis
- Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA.
| | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
47
|
Avin A, Levy M, Porat Z, Abramson J. Quantitative analysis of protein-protein interactions and post-translational modifications in rare immune populations. Nat Commun 2017; 8:1524. [PMID: 29142256 PMCID: PMC5688095 DOI: 10.1038/s41467-017-01808-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/17/2017] [Indexed: 01/23/2023] Open
Abstract
In spite of recent advances in proteomics, quantitative analyses of protein-protein interactions (PPIs) or post-translational modifications (PTMs) in rare cell populations remain challenging. This is in particular true for analyses of rare immune and/or stem cell populations that are directly isolated from humans or animal models, and which are often characterized by multiple surface markers. To overcome these limitations, here we have developed proximity ligation imaging cytometry (PLIC), a protocol for proteomic analysis of rare cells. Specifically, by employing PLIC on medullary thymic epithelial cells (mTECs), which serve as a paradigm for a rare immune population, we demonstrate that PLIC overcomes the inherent limitations of conventional proteomic approaches and enables a high-resolution detection and quantification of PPIs and PTMs at a single cell level.
Collapse
Affiliation(s)
- Ayelet Avin
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Maayan Levy
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Ziv Porat
- Department of Biological Services, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Jakub Abramson
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
48
|
Abstract
Intrathymic T cell development is a complex process that depends upon continuous guidance from thymus stromal cell microenvironments. The thymic epithelium within the thymic stroma comprises highly specialized cells with a high degree of anatomic, phenotypic, and functional heterogeneity. These properties are collectively required to bias thymocyte development toward production of self-tolerant and functionally competent T cells. The importance of thymic epithelial cells (TECs) is evidenced by clear links between their dysfunction and multiple diseases where autoimmunity and immunodeficiency are major components. Consequently, TECs are an attractive target for cell therapies to restore effective immune system function. The pathways and molecular regulators that control TEC development are becoming clearer, as are their influences on particular stages of T cell development. Here, we review both historical and the most recent advances in our understanding of the cellular and molecular mechanisms controlling TEC development, function, dysfunction, and regeneration.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Graham Anderson
- MRC Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
49
|
Reversible modulation of SIRT1 activity in a mouse strain. PLoS One 2017; 12:e0173002. [PMID: 28273169 PMCID: PMC5342236 DOI: 10.1371/journal.pone.0173002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/13/2017] [Indexed: 12/18/2022] Open
Abstract
The SIRT1 protein deacetylase is reported to have a remarkably wide spectrum of biological functions affecting such varied processes as aging, cancer, metabolism, neurodegeneration and immunity. However, the SIRT1 literature is also full of contradictions. To help establish the role(s) of SIRT1 in these and other biological processes, we set out to create a mouse in which the SIRT1 activity could be toggled between on and off states by fusing the estrogen receptor ligand-binding domain (ER) to the C terminus of the SIRT1 protein. We found that the catalytic activity of the SIRT1-ER fusion protein increased 4–5 fold in cells treated with its ligand, 4-hydroxy-tamoxifen (4OHT). The 4OHT-induced activation of SIRT1-ER was due in large part to a 2 to 4-fold increase in abundance of the SIRT1-ER protein in cells in culture and in tissues in vivo. This increase is reversible and is a consequence of 4OHT-induced stabilization of the SIRT1-ER protein. Since changes in SIRT1 level or activity of 2–4 fold are frequently reported to be sufficient to affect its biological functions, this mouse should be helpful in establishing the causal relationships between SIRT1 and the diseases and processes it affects.
Collapse
|
50
|
Souslova T, Mirédin K, Millar AM, Albert PR. Recruitment by the Repressor Freud-1 of Histone Deacetylase-Brg1 Chromatin Remodeling Complexes to Strengthen HTR1A Gene Repression. Mol Neurobiol 2016; 54:8263-8277. [PMID: 27914010 DOI: 10.1007/s12035-016-0306-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/16/2016] [Indexed: 12/12/2022]
Abstract
Five-prime repressor element under dual repression binding protein-1 (Freud-1)/CC2D1A is genetically linked to intellectual disability and implicated in neuronal development. Freud-1 represses the serotonin-1A (5-HT1A) receptor gene HTR1A by histone deacetylase (HDAC)-dependent or HDAC-independent mechanisms in 5-HT1A-negative (e.g., HEK-293) or 5-HT1A-expressing cells (SK-N-SH), respectively. To identify the underlying mechanisms, Freud-1-associated proteins were affinity-purified from HEK-293 nuclear extracts and members of the Brg1/SMARCCA chromatin remodeling and Sin3A-HDAC corepressor complexes were identified. Pull-down assays using recombinant proteins showed that Freud-1 interacts directly with the Brg1 carboxyl-terminal domain; interaction with Brg1 required the carboxyl-terminal of Freud-1. Freud-1 complexes in HEK-293 and SK-N-SH cells differed, with low levels of BAF170/SMARCC2 and BAF57/SMARCE1 in HEK-293 cells and low-undetectable BAF155/SMARCC1, Sin3A, and HDAC1/2 in SK-N-SH cells. Similarly, by quantitative chromatin immunoprecipitation, Brg1-BAF170/57 and Sin3A-HDAC complexes were observed at the HTR1A promoter in HEK-293 cells, whereas in SK-N-SH cells, Sin3A-HDAC proteins were not detected. Quantifying 5-HT1A receptor mRNA levels in cells treated with siRNA to Freud-1, Brg1, or both RNAs addressed the functional role of the Freud-1-Brg1 complex. In HEK-293 cells, 5-HT1A receptor mRNA levels were increased only when both Freud-1 and Brg1 were depleted, but in SK-N-SH cells, depletion of either protein upregulated 5-HT1A receptor RNA. Thus, recruitment by Freud-1 of Brg1, BAF155, and Sin3A-HDAC complexes appears to strengthen repression of the HTR1A gene to prevent its expression inappropriate cell types, while recruitment of the Brg1-BAF170/57 complex is permissive to 5-HT1A receptor expression. Alterations in Freud-1-Brg1 interactions in mutants associated with intellectual disability could impair gene repression leading to altered neuronal development.
Collapse
Affiliation(s)
- Tatiana Souslova
- Ottawa Hospital Research Institute (Neuroscience) and UOttawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Kim Mirédin
- Ottawa Hospital Research Institute (Neuroscience) and UOttawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Anne M Millar
- Ottawa Hospital Research Institute (Neuroscience) and UOttawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience) and UOttawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|