1
|
Kucuka I, Iraji D, Braun S, Breivik L, Wolff ASB, Husebye ES, Oftedal BE. Longitudinal Immune Profiling in Autoimmune Polyendocrine Syndrome Type 1. Scand J Immunol 2025; 101:e70021. [PMID: 40166896 PMCID: PMC11959528 DOI: 10.1111/sji.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 02/24/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
Autoimmune polyendocrine syndrome Type-1 (APS-1) is a rare, but severe organ-specific autoimmune disease caused by mutations in the autoimmune regulator (AIRE) gene. Lack of AIRE causes autoreactive T cells to escape negative selection and alters the T regulatory cell subset. However, little is known about how the immune cell subsets vary across the lifespan in APS-1. Here we analysed the peripheral distribution of 13 immune cell subsets along the lifespan using epigenetic quantification. We found the largest discrepancy in immune cells to appear early in APS-1 patients' lives, coinciding with the time point they obtained most of their clinical symptoms. We further revealed longitudinal changes in cell compositions both within the adaptive and the innate arms of the immune system. We found that cell frequencies of B cells, T-cell subgroups, nonclassical monocytes, and Natural Killer cells to be reduced in young APS-1 patients. We also found B-cell frequencies to decrease with ageing in both patients and healthy controls. Our results suggest that Tregs, follicular helper T, and natural killer cells have opposing trends of cell frequencies during life, indicating the importance of considering the age profiles of cohorts which could otherwise lead to conflicting conclusions.
Collapse
Affiliation(s)
- Isil Kucuka
- Department of Clinical Science, Department of MedicineUniversity of BergenBergenNorway
| | - Dorsa Iraji
- Department of Clinical Science, Department of MedicineUniversity of BergenBergenNorway
| | - Sarah Braun
- Department of Clinical Science, Department of MedicineUniversity of BergenBergenNorway
| | - Lars Breivik
- Department of Clinical Science, Department of MedicineUniversity of BergenBergenNorway
- Department of MedicineHaukeland University HospitalBergenNorway
| | - Anette S. B. Wolff
- Department of Clinical Science, Department of MedicineUniversity of BergenBergenNorway
- Department of MedicineHaukeland University HospitalBergenNorway
- Health Research Sogn Og Fjordane, Førde Hospital TrustFørdeNorway
| | - Eystein S. Husebye
- Department of Clinical Science, Department of MedicineUniversity of BergenBergenNorway
- Department of MedicineHaukeland University HospitalBergenNorway
| | - Bergithe E. Oftedal
- Department of Clinical Science, Department of MedicineUniversity of BergenBergenNorway
| |
Collapse
|
2
|
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
|
3
|
Xu Y, Chen W, Wu X, Zhao K, Liu C, Zhang H. The Role of Cells and Cytokines in Male Infertility Induced by Orchitis. World J Mens Health 2024; 42:681-693. [PMID: 38449458 PMCID: PMC11439807 DOI: 10.5534/wjmh.230270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/05/2023] [Accepted: 11/19/2023] [Indexed: 03/08/2024] Open
Abstract
Recent studies on male infertility reveal a growing worry: more infertile men are dealing with inflammation in the testis. Analyzing testicular biopsies from infertile men highlights a significant presence of inflammation. This connection, supported by clinical and pathological evidence, emphasizes that testicular inflammation hampers sperm production, leading to lasting declines in sperm count and quality. However, the exact reasons behind male infertility due to orchitis, a type of testicular inflammation, are still uncertain. Understanding these fundamental aspects of molecular signals and cellular mechanisms in testicular inflammation is crucial. Our review delves into recent literature with a dual objective: elucidating potential mechanisms involving immune cells, non-immune cells, and cytokines that link orchitis to male infertility, while also paving the way for precise interventions and solutions to address the challenges of male infertility.
Collapse
Affiliation(s)
- Ying Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyi Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Wu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Hetemäki I, Sarkkinen J, Heikkilä N, Drechsel K, Mäyränpää MI, Färkkilä A, Laakso S, Mäkitie O, Arstila TP, Kekäläinen E. Dysregulated germinal center reaction with expanded T follicular helper cells in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy lymph nodes. J Allergy Clin Immunol 2024; 153:1445-1455. [PMID: 38128835 DOI: 10.1016/j.jaci.2023.12.004] [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] [Received: 07/05/2023] [Revised: 10/30/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED, also called APS-1) is an inborn error of immunity with clear signs of B-cell autoimmunity such as neutralizing anti-IFN antibodies. In APECED, mutations in the AIRE gene impair thymic negative selection of T cells. The resulting T-cell alterations may then cause dysregulation of B-cell responses. However, no analysis of interactions of T and B cells in the germinal centers (GCs) in patients' secondary lymphatic tissues has been reported. OBJECTIVE This study examined the relationship between B cells and follicular T helper cells (TfH) in peripheral blood and lymph node (LN) GCs in patients with APECED. METHODS Immunophenotyping of peripheral blood B cells and TfH was performed for 24 patients with APECED. Highly multiplexed fluorescent immunohistochemical staining was performed on 7 LN biopsy samples from the patients to study spatial interactions of lymphocytes in the GCs at the single-cell level. RESULTS The patients' peripheral B-cell phenotype revealed skewing toward a mature B-cell phenotype with marked loss of transitional and naive B cells. The frequency of circulating TfH cells was diminished in the patients, while in the LNs the TfH population was expanded. In LNs the overall frequency of Treg cells and interactions of Treg cells with nonfollicular T cells were reduced, suggesting that aberrant Treg cell function might fail to restrain TfH differentiation. CONCLUSIONS GC reactions are disrupted in APECED as a result of defective T-cell control.
Collapse
Affiliation(s)
- Iivo Hetemäki
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - Joona Sarkkinen
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Nelli Heikkilä
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Karen Drechsel
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko I Mäyränpää
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anniina Färkkilä
- Research Program in Systems Oncology, FIMM & HiLIFE University of Helsinki, Helsinki, Finland; iCAN Digital Precision Cancer Medicine, Helsinki, Finland; Department of Obstetrics and Gynecology, University Hospital, Helsinki, Finland
| | - Saila Laakso
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
| | - Outi Mäkitie
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Department of Molecular Medicine, Karolinska Institutet, and Clinical Genetics, Karolinska University Hospital, Stockholm, Stockholm, Sweden
| | - T Petteri Arstila
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eliisa Kekäläinen
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
5
|
Sjøgren T, Islam S, Filippov I, Jebrzycka A, Sulen A, Breivik LE, Hellesen A, Jørgensen AP, Lima K, Tserel L, Kisand K, Peterson P, Ranki A, Husebye ES, Oftedal BE, Wolff AS. Single cell characterization of blood and expanded regulatory T cells in autoimmune polyendocrine syndrome type 1. iScience 2024; 27:109610. [PMID: 38632993 PMCID: PMC11022049 DOI: 10.1016/j.isci.2024.109610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
Immune tolerance fails in autoimmune polyendocrine syndrome type 1 (APS-1) because of AIRE mutations. We have used single cell transcriptomics to characterize regulatory T cells (Tregs) sorted directly from blood and from in vitro expanded Tregs in APS-1 patients compared to healthy controls. We revealed only CD52 and LTB (down) and TXNIP (up) as consistently differentially expressed genes in the datasets. There were furthermore no large differences of the TCR-repertoire of expanded Tregs between the cohorts, but unique patients showed a more restricted use of specific clonotypes. We also found that in vitro expanded Tregs from APS-1 patients had similar suppressive capacity as controls in co-culture assays, despite expanding faster and having more exhausted cells. Our results suggest that APS-1 patients do not have intrinsic defects in their Treg functionality, and that their Tregs can be expanded ex vivo for potential therapeutic applications.
Collapse
Affiliation(s)
- Thea Sjøgren
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Shahinul Islam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Igor Filippov
- QIAGEN Aarhus A/S, Aarhus, Denmark
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | | | - André Sulen
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars E. Breivik
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | | | | | - Kari Lima
- Department of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Liina Tserel
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereology, University of Helsinki and Helsinki University Hospital, Inflammation Centre, Helsinki, Finland
| | - Eystein S. Husebye
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Bergithe E. Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S.B. Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
6
|
Oftedal BE, Sjøgren T, Wolff ASB. Interferon autoantibodies as signals of a sick thymus. Front Immunol 2024; 15:1327784. [PMID: 38455040 PMCID: PMC10917889 DOI: 10.3389/fimmu.2024.1327784] [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: 10/25/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Type I interferons (IFN-I) are key immune messenger molecules that play an important role in viral defense. They act as a bridge between microbe sensing, immune function magnitude, and adaptive immunity to fight infections, and they must therefore be tightly regulated. It has become increasingly evident that thymic irregularities and mutations in immune genes affecting thymic tolerance can lead to the production of IFN-I autoantibodies (autoAbs). Whether these biomarkers affect the immune system or tissue integrity of the host is still controversial, but new data show that IFN-I autoAbs may increase susceptibility to severe disease caused by certain viruses, including SARS-CoV-2, herpes zoster, and varicella pneumonia. In this article, we will elaborate on disorders that have been identified with IFN-I autoAbs, discuss models of how tolerance to IFN-Is is lost, and explain the consequences for the host.
Collapse
Affiliation(s)
- Bergithe E. Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Thea Sjøgren
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S. B. Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
7
|
Sjøgren T, Bjune JI, Husebye ES, Oftedal BE, Wolff ASB. Regulatory T cells in autoimmune primary adrenal insufficiency. Clin Exp Immunol 2024; 215:47-57. [PMID: 37578839 PMCID: PMC10776243 DOI: 10.1093/cei/uxad087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/01/2023] [Accepted: 08/11/2023] [Indexed: 08/15/2023] Open
Abstract
Primary adrenal insufficiency (PAI) is most often caused by an autoimmune destruction of the adrenal cortex resulting in failure to produce cortisol and aldosterone. The aetiology is thought to be a combination of genetic and environmental risk factors, leading to breakdown of immunological tolerance. Regulatory T cells (Tregs) are deficient in many autoimmune disorders, but it is not known whether they contribute to development of PAI. We aimed to investigate the frequency and function of naive and expanded Tregs in patients with PAI and polyendocrine syndromes compared to age- and gender-matched healthy controls. Flow cytometry was used to assess the frequency and characterize functional markers of blood Tregs in PAI (N = 15). Expanded Treg suppressive abilities were assessed with a flow cytometry based suppression assay (N = 20), while bulk RNA-sequencing was used to examine transcriptomic differences (N = 16) and oxygen consumption rate was measured by a Seahorse cell metabolic assay (N = 11). Our results showed that Treg frequency and suppressive capacity were similar between patients and controls. An increased expression of killer-cell leptin-like receptors and mitochondrial genes was revealed in PAI patients, but their expanded Tregs did not display signs of mitochondrial dysfunction. Our findings do not support a clear role for Tregs in the contribution of PAI development.
Collapse
Affiliation(s)
- Thea Sjøgren
- Endocrine Medicine Group, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jan-Inge Bjune
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Eystein S Husebye
- Endocrine Medicine Group, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Bergithe E Oftedal
- Endocrine Medicine Group, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S B Wolff
- Endocrine Medicine Group, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
8
|
Peterson P. Novel Insights into the Autoimmunity from the Genetic Approach of the Human Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:3-18. [PMID: 38467969 DOI: 10.1007/978-981-99-9781-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Autoimmune-polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a monogenic inborn error of autoimmunity that is caused by damaging germline variants in the AIRE gene and clinically manifests with multiple autoimmune diseases in patients. Studies on the function of the AIRE gene, discovered in 1997, have contributed to fundamental aspects of human immunology as they have been important in understanding the basic mechanism of immune balance between self and non-self. This chapter looks back to the discovery of the AIRE gene, reviews its main properties, and discusses the key findings of its function in the thymus. However, more recent autoantibody profilings in APECED patients have highlighted a gap in our knowledge of the disease pathology and point to the need to revisit the current paradigm of AIRE function. The chapter reviews these new findings in APECED patients, which potentially trigger new thoughts on the mechanism of immune tolerance.
Collapse
Affiliation(s)
- Pärt Peterson
- Institute of Biomedical and Translational Medicine, University of Tartu, Tartu, Estonia.
| |
Collapse
|
9
|
Ni D, Tan J, Robert R, Taitz J, Ge A, Potier-Villette C, Reyes JGA, Spiteri A, Wishart C, Mackay C, Piccio L, King NJC, Macia L. GPR109A expressed on medullary thymic epithelial cells affects thymic Treg development. Eur J Immunol 2023; 53:e2350521. [PMID: 37595951 DOI: 10.1002/eji.202350521] [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] [Received: 04/06/2023] [Revised: 07/31/2023] [Accepted: 08/17/2023] [Indexed: 08/20/2023]
Abstract
Regulatory T cells (Treg) maintain immune homeostasis due to their anti-inflammatory functions. They can be generated either centrally in the thymus or in peripheral organs. Metabolites such as short-chain fatty acids produced by intestinal microbiota can induce peripheral Treg differentiation, by activating G-protein-coupled-receptors like GPR109A. In this study, we identified a novel role for GPR109A in thymic Treg development. We found that Gpr109a-/- mice had increased Treg under basal conditions in multiple organs compared with WT mice. GPR109A was not expressed on T cells but on medullary thymic epithelial cells (mTECs), as revealed by single-cell RNA sequencing in both mice and humans and confirmed by flow cytometry in mice. mTECs isolated from Gpr109a-/- mice had higher expression of autoimmune regulator (AIRE), the key regulator of Treg development, while the subset of mTECs that did not express Gpr109a in the WT displayed increased Aire expression and also enhanced signaling related to mTEC functionality. Increased thymic Treg in Gpr109a-/- mice was associated with protection from experimental autoimmune encephalomyelitis, with ameliorated clinical signs and reduced inflammation. This work identifies a novel role for GPR109A and possibly the gut microbiota, on thymic Treg development via its regulation of mTECs.
Collapse
Affiliation(s)
- Duan Ni
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Jian Tan
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Remy Robert
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jemma Taitz
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anjie Ge
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Camille Potier-Villette
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Julen Gabirel Araneta Reyes
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alanna Spiteri
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, The School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Claire Wishart
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, The School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Charles Mackay
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Laura Piccio
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicholas Jonathan Cole King
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, The School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, New South Wales, Australia
| |
Collapse
|
10
|
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
|
11
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 316] [Impact Index Per Article: 158.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
12
|
Almaghrabi S, Lovewell T, Azzouz M, Tazi Ahnini R. Characterisation of APS-1 Experimental Models Is Crucial for Development of Novel Therapies. BIOMED RESEARCH INTERNATIONAL 2023; 2023:7960443. [PMID: 36685668 PMCID: PMC9848810 DOI: 10.1155/2023/7960443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 11/02/2022] [Accepted: 12/13/2022] [Indexed: 01/13/2023]
Abstract
Autoimmune polyglandular syndrome type 1 (APS-1) is an inherited autosomal disorder. The most common clinical features of the disease include adrenocortical failure, hypoparathyroidism (HP), and chronic mucocutaneous candidiasis (CMC). APS-1 is caused by mutations in the autoimmune regulator (AIRE) gene. AIRE is a transcriptional factor involved in the regulation of thousands of genes in the thymus. It facilitates central tolerance by promoting the ectopic expression of tissue-specific antigens (TSAs) in medullary thymic epithelial cells (mTECs), leading to the deletion of self-reactive thymocytes. Several Aire-deficient mice were developed separately, on different backgrounds; seven published Aire knockout mice show a variety of phenotypes depending on the strain used to generate the experimental model. The first Aire-deficient mice were generated on a "black 6" background almost 20 years ago. The model showed mild phenotype with relatively modest penetrance compared to models generated on BALBc or NOD backgrounds. The generation of all these experimental models is crucial for development and testing new therapeutics as well as reading the response to treatments.
Collapse
Affiliation(s)
- Sarah Almaghrabi
- Department of Infection, Immunity and Cardiovascular Disease, Sheffield, UK
| | - Thomas Lovewell
- Department of Infection, Immunity and Cardiovascular Disease, Sheffield, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience (SITRaN), Department of Neuroscience, The Medical School, University of Sheffield, Sheffield S10 2RX, UK
| | - Rachid Tazi Ahnini
- Department of Infection, Immunity and Cardiovascular Disease, Sheffield, UK
| |
Collapse
|
13
|
Santacroce L, Imbimbo C, Ballini A, Crocetto F, Scacco S, Cantore S, Di Zazzo E, Colella M, Jirillo E. Testicular Immunity and Its Connection with the Microbiota. Physiological and Clinical Implications in the Light of Personalized Medicine. J Pers Med 2022; 12:1335. [PMID: 36013286 PMCID: PMC9409709 DOI: 10.3390/jpm12081335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 12/03/2022] Open
Abstract
Reproduction is a complex process, which is based on the cooperation between the endocrine-immune system and the microbiota. Testicular immunity is characterized by the so-called immune privilege, a mechanism that avoids autoimmune attacks against proteins expressed by spermatozoa. Testicular microbiota is connected with the gut microbiota, the most prevalent site of commensals inthe body. Both microbiotas take part inthe development of the immune system and protection againstpathogen invasion. Dysbiosis is caused by concurrent pathologies, such as obesity, diabetes, infections and trauma. The substitution of beneficial bacteria with pathogens may lead to destruction of spermatozoa directly or indirectly and, ultimately, to male infertility. Novel therapeutic interventions, i.e., nutritional interventions and supplementation of natural products, such as, probiotics, prebiotics, antioxidants and polyphenols, may lead to the restoration of the otherwise-impaired male reproductive potential, even if experimental and clinical results are not always concordant. In this review, the structure and immune function of the testis will be described with special reference to the blood-testisbarrier. The regulatory role of both the gut and testicular microbiota will be illustrated in health and disease, also emphasizing therapeutic attempts with natural products for the correction of male infertility, in the era of personalized medicine.
Collapse
Affiliation(s)
- Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Ciro Imbimbo
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131Naples, Italy
| | - Andrea Ballini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Felice Crocetto
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131Naples, Italy
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Stefania Cantore
- Independent Researcher, Sorriso & Benessere—Ricerca e Clinica, 70129 Bari, Italy
| | - Erika Di Zazzo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Emilio Jirillo
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| |
Collapse
|
14
|
Charaix J, Borelli A, Santamaria JC, Chasson L, Giraud M, Sergé A, Irla M. Recirculating Foxp3 + regulatory T cells are restimulated in the thymus under Aire control. Cell Mol Life Sci 2022; 79:355. [PMID: 35678896 PMCID: PMC11071703 DOI: 10.1007/s00018-022-04328-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023]
Abstract
Thymically-derived Foxp3+ regulatory T cells (Treg) critically control immunological tolerance. These cells are generated in the medulla through high affinity interactions with medullary thymic epithelial cells (mTEC) expressing the Autoimmune regulator (Aire). Recent advances have revealed that thymic Treg contain not only developing but also recirculating cells from the periphery. Although Aire is implicated in the generation of Foxp3+ Treg, its role in the biology of recirculating Treg remains elusive. Here, we show that Aire regulates the suppressive signature of recirculating Treg independently of the remodeling of the medullary 3D organization throughout life where Treg reside. Accordingly, the adoptive transfer of peripheral Foxp3+ Treg in AireKO recipients led to an impaired suppressive signature upon their entry into the thymus. Furthermore, recirculating Treg from AireKO mice failed to attenuate the severity of multiorgan autoimmunity, demonstrating that their suppressive function is altered. Using bone marrow chimeras, we reveal that mTEC-specific expression of Aire controls the suppressive signature of recirculating Treg. Finally, mature mTEC lacking Aire were inefficient in stimulating peripheral Treg both in polyclonal and antigen-specific co-culture assays. Overall, this study demonstrates that Aire confers to mTEC the ability to restimulate recirculating Treg, unravelling a novel function for this master regulator in Treg biology.
Collapse
Affiliation(s)
- Jonathan Charaix
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Alexia Borelli
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Jérémy C Santamaria
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Lionel Chasson
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Matthieu Giraud
- Center for Research in Transplantation and Translational Immunology, UMR 1064, INSERM, Nantes Université, 44000, Nantes, France
| | - Arnauld Sergé
- Turing Centre for Living Systems, Laboratoire adhésion inflammation (LAI), CNRS, INSERM, Aix-Marseille University, 13288, Marseille, France
| | - Magali Irla
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
15
|
Fierabracci A, Belcastro E, Carbone E, Pagliarosi O, Palma A, Pacillo L, Giancotta C, Zangari P, Finocchi A, Cancrini C, Delfino DV, Cappa M, Betterle C. In Search for the Missing Link in APECED-like Conditions: Analysis of the AIRE Gene in a Series of 48 Patients. J Clin Med 2022; 11:3242. [PMID: 35683627 PMCID: PMC9181695 DOI: 10.3390/jcm11113242] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/20/2022] [Accepted: 05/30/2022] [Indexed: 11/26/2022] Open
Abstract
Autoimmune diseases are a heterogeneous group of disorders of the immune system. They can cluster in the same individual, revealing various preferential associations for polyendocrine autoimmune syndromes. Clinical observation, together with advances in genetics and the understanding of pathophysiological processes, has further highlighted that autoimmunity can be associated with immunodeficiency; autoimmunity may even be the first primary immunodeficiency manifestation. Analysis of susceptibility genes for the development of these complex phenotypes is a fundamental issue. In this manuscript, we revised the clinical and immunologic features and the presence of AIRE gene variations in a cohort of 48 patients affected by high polyautoimmunity complexity, i.e., APECED-like conditions, also including patients affected by primary immunodeficiency. Our results evidenced a significant association of the S278R polymorphism of the AIRE gene with APECED-like conditions, including both patients affected by autoimmunity and immunodeficiency and patients with polyautoimmunity compared to healthy controls. A trend of association was also observed with the IVS9+6 G>A polymorphism. The results of this genetic analysis emphasize the need to look for additional genetic determinants playing in concert with AIRE polymorphisms. This will help to improve the diagnostic workup and ensure a precision medicine approach to targeted therapies in APECED-like patients.
Collapse
Affiliation(s)
- Alessandra Fierabracci
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), 00146 Rome, Italy; (E.B.); (E.C.); (O.P.)
| | - Eugenia Belcastro
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), 00146 Rome, Italy; (E.B.); (E.C.); (O.P.)
| | - Elena Carbone
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), 00146 Rome, Italy; (E.B.); (E.C.); (O.P.)
| | - Olivia Pagliarosi
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), 00146 Rome, Italy; (E.B.); (E.C.); (O.P.)
| | - Alessia Palma
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Lucia Pacillo
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.P.); (A.F.); (C.C.)
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Carmela Giancotta
- Immunology and Vaccinology, DPUO, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.G.); (P.Z.)
| | - Paola Zangari
- Immunology and Vaccinology, DPUO, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.G.); (P.Z.)
| | - Andrea Finocchi
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.P.); (A.F.); (C.C.)
- Chair of Pediatrics, Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Caterina Cancrini
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.P.); (A.F.); (C.C.)
- Chair of Pediatrics, Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Marco Cappa
- Endocrinology Unit, DPUO, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Corrado Betterle
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy;
| |
Collapse
|
16
|
Skrabic V, Skrabic I, Skrabic R, Roje B, Simunovic M. Clinical Characteristics in the Longitudinal Follow-Up of APECED Syndrome in Southern Croatia—Case Series. Genes (Basel) 2022; 13:genes13040558. [PMID: 35456364 PMCID: PMC9027969 DOI: 10.3390/genes13040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/11/2022] [Accepted: 03/19/2022] [Indexed: 02/03/2023] Open
Abstract
Background: Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED) is a rare monogenetic autosomal recessive disorder caused by a mutation in the autoimmune regulator (AIRE) gene characterized by complex phenotypic characteristics discovered over years of follow-up. Methods: 7 patients were recruited in this case series in a period of the last 37 years from Southern Croatia. All patients were screened for AIRE R257X mutations. Results: This study group had a mean current age of 25.3 years (age range from 5.4 to 40.2 years), while the mean age at the onset of the disease was 6.5 years (age range from 0.7 to 9.2 years) and with a mean follow-up period of 17.8 years. The overall prevalence of APECED syndrome is estimated to be 1 in 75,000. The most common initial manifestation of the disease was onychodystrophy, while the first major component of APECED syndrome was chronic mucocutaneous candidiasis. Conclusions: APECED is a ‘‘multi-faced’’ disease based on the very unpredictable and inconsistent onset of major components. Furthermore, based on our results, we suggest that onychodystrophy could be included as a warning sign of APECED syndrome.
Collapse
Affiliation(s)
- Veselin Skrabic
- Department of Pediatrics, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia; (I.S.); (M.S.)
- Department of Pediatrics, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
- Correspondence:
| | - Ivna Skrabic
- Department of Pediatrics, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia; (I.S.); (M.S.)
| | - Roko Skrabic
- Department of Nephrology, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia;
| | - Blanka Roje
- Laboratory for Cancer Research, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia;
| | - Marko Simunovic
- Department of Pediatrics, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia; (I.S.); (M.S.)
- Department of Pediatrics, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| |
Collapse
|
17
|
Padonou F, Gonzalez V, Provin N, Yayilkan S, Jmari N, Maslovskaja J, Kisand K, Peterson P, Irla M, Giraud M. Aire-dependent transcripts escape Raver2-induced splice-event inclusion in the thymic epithelium. EMBO Rep 2022; 23:e53576. [PMID: 35037357 PMCID: PMC8892270 DOI: 10.15252/embr.202153576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/14/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022] Open
Abstract
Aire allows medullary thymic epithelial cells (mTECs) to express and present a large number of self-antigens for central tolerance. Although mTECs express a high diversity of self-antigen splice isoforms, the extent and regulation of alternative splicing events (ASEs) in their transcripts, notably in those induced by Aire, is unknown. In contrast to Aire-neutral genes, we find that transcripts of Aire-sensitive genes show only a low number of ASEs in mTECs, with about a quarter present in peripheral tissues excluded from the thymus. We identify Raver2, as a splicing-related factor overexpressed in mTECs and dependent on H3K36me3 marks, that promotes ASEs in transcripts of Aire-neutral genes, leaving Aire-sensitive ones unaffected. H3K36me3 profiling reveals its depletion at Aire-sensitive genes and supports a mechanism that is preceding Aire expression leading to transcripts of Aire-sensitive genes with low ASEs that escape Raver2-induced alternative splicing. The lack of ASEs in Aire-induced transcripts would result in an incomplete Aire-dependent negative selection of autoreactive T cells, thus highlighting the need of complementary tolerance mechanisms to prevent activation of these cells in the periphery.
Collapse
Affiliation(s)
- Francine Padonou
- Nantes UniversitéINSERMCenter for Research in Transplantation and Translational Immunology, UMR 1064NantesFrance,Institut CochinINSERMCNRSParis UniversitéParisFrance
| | | | - Nathan Provin
- Nantes UniversitéINSERMCenter for Research in Transplantation and Translational Immunology, UMR 1064NantesFrance
| | - Sümeyye Yayilkan
- Nantes UniversitéINSERMCenter for Research in Transplantation and Translational Immunology, UMR 1064NantesFrance
| | - Nada Jmari
- Institut CochinINSERMCNRSParis UniversitéParisFrance
| | | | - Kai Kisand
- Molecular Pathology Research GroupUniversity of TartuTartuEstonia
| | - Pärt Peterson
- Molecular Pathology Research GroupUniversity of TartuTartuEstonia
| | - Magali Irla
- Aix‐Marseille UniversitéCNRSINSERMCIML, Centre d'Immunologie de Marseille‐LuminyMarseilleFrance
| | - Matthieu Giraud
- Nantes UniversitéINSERMCenter for Research in Transplantation and Translational Immunology, UMR 1064NantesFrance,Institut CochinINSERMCNRSParis UniversitéParisFrance
| |
Collapse
|
18
|
Besnard M, Sérazin C, Ossart J, Moreau A, Vimond N, Flippe L, Sein H, Smith GA, Pittaluga S, Ferré EM, Usal C, Anegon I, Ranki A, Lionakis MS, Peterson P, Guillonneau C. Anti-CD45RC antibody immunotherapy prevents and treats experimental Autoimmune PolyEndocrinopathy Candidiasis Ectodermal Dystrophy syndrome. J Clin Invest 2022; 132:156507. [PMID: 35167497 PMCID: PMC8970675 DOI: 10.1172/jci156507] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
Targeted monoclonal antibody (mAb) therapies show great promise for the treatment of transplant rejection and autoimmune diseases by inducing more specific immunomodulatory effects than broadly immunosuppressive drugs routinely used. We recently described the therapeutic advantage of targeting CD45RC, expressed at high levels by conventional T cells (Tconv, CD45RChigh), their precursors and terminally differentiated T (TEMRA) cells, but not by regulatory T cells (Tregs, CD45RClow/-). We demonstrated efficacy of anti-CD45RC mAb treatment in transplantation but its potential has not been examined in autoimmune diseases. APECED is a rare genetic syndrome caused by loss-of-function mutations of the key central tolerance mediator, autoimmune regulator (AIRE) leading to abnormal auto-reactive T cell responses and autoantibodies production. Herein, we showed that, in a rat model of APECED syndrome, anti-CD45RC mAb was effective both as prevention and treatment of autoimmune manifestations and inhibited autoantibody development. Anti-CD45RC mAb intervention depleted CD45RChigh T cells, inhibited CD45RChigh B cells, and restored the Treg/Tconv ratio and the altered Tregs transcriptomic profile. In APECED patients, CD45RC was significantly increased in peripheral blood T cells and lesioned organs from APECED patients were infiltrated by CD45RChigh cells. Our observations highlight the potential role for CD45RChigh cells in the pathogenesis of experimental and human APECED syndrome and the potential of anti-CD45RC antibody treatment.
Collapse
Affiliation(s)
- Marine Besnard
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, University of Nantes, Nantes, France
| | - Céline Sérazin
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, University of Nantes, Nantes, France
| | - Jason Ossart
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, University of Nantes, Nantes, France
| | - Anne Moreau
- Department of Pathology, CHU Nantes, Nantes, France
| | - Nadège Vimond
- Department of Immunology, AbolerIS Pharma, Nantes, France
| | - Léa Flippe
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, University of Nantes, Nantes, France
| | - Hanna Sein
- Department of Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Grace A Smith
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States of America
| | | | - Elise Mn Ferré
- Laboratory of Clinical Immunology and Microbiology, NIAID/NIH, Bethesda, United States of America
| | - Claire Usal
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, University of Nantes, Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, University of Nantes, Nantes, France
| | - Annamari Ranki
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology, NIAID/NIH, Bethesda, United States of America
| | - Pärt Peterson
- Department of Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, University of Nantes, Nantes, France
| |
Collapse
|
19
|
Morimoto J, Matsumoto M, Miyazawa R, Yoshida H, Tsuneyama K, Matsumoto M. Aire suppresses CTLA-4 expression from the thymic stroma to control autoimmunity. Cell Rep 2022; 38:110384. [PMID: 35172142 DOI: 10.1016/j.celrep.2022.110384] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/08/2021] [Accepted: 01/24/2022] [Indexed: 01/07/2023] Open
Abstract
Impaired production of thymic regulatory T cells (Tregs) is implicated in the development of Aire-dependent autoimmunity. Because Tregs require agonistic T cell receptor stimuli by self-antigens to develop, reduced expression of self-antigens from medullary thymic epithelial cells (mTECs) has been considered to play a major role in the reduced Treg production in Aire deficiency. Here, we show that mTECs abnormally express co-inhibitory receptor CTLA-4 if Aire is non-functional. Upon binding with CD80/CD86 ligands expressed on thymic dendritic cells (DCs), the ectopically expressed CTLA-4 from Aire-deficient mTECs removes the CD80/CD86 ligands from the DCs. This attenuates the ability of DCs to provide co-stimulatory signals and to present self-antigens transferred from mTECs, both of which are required for Treg production. Accordingly, impaired production of Tregs and organ-specific autoimmunity in Aire-deficient mice are rescued by the depletion of CTLA-4 expression from mTECs. Our studies illuminate the significance of mTEC-DC interaction coordinated by Aire for the establishment of thymic tolerance.
Collapse
Affiliation(s)
- 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 Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| | - Ryuichiro Miyazawa
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Hideyuki Yoshida
- YCI Laboratory for Immunological Transcriptomics, RIKEN Center for Integrative Medical Science, Yokohama 230-0045, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan.
| |
Collapse
|
20
|
Laakso S, Holopainen E, Betterle C, Saari V, Vogt E, Schmitt MM, Winer KK, Kareva M, Sabbadin C, Husebye ES, Orlova E, Lionakis MS, Mäkitie O. Pregnancy Outcome in Women With APECED (APS-1): A Multicenter Study on 43 Females With 83 Pregnancies. J Clin Endocrinol Metab 2022; 107:e528-e537. [PMID: 34570215 PMCID: PMC8764323 DOI: 10.1210/clinem/dgab705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Indexed: 01/19/2023]
Abstract
CONTEXT Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED; also known as autoimmune polyendocrine syndrome type 1) has a severe, unpredictable course. Autoimmunity and disease components may affect fertility and predispose to maternal and fetal complications, but pregnancy outcomes remain unknown. OBJECTIVE To assess fetal and maternal outcomes and course of clinical APECED manifestations during pregnancy in women with APECED. DESIGN AND SETTING A multicenter registry-based study including 5 national patient cohorts. PATIENTS 321 females with APECED. MAIN OUTCOME MEASURE Number of pregnancies, miscarriages, and deliveries. RESULTS Forty-three patients had altogether 83 pregnancies at median age of 27 years (range, 17-39). Sixty (72%) pregnancies led to a delivery, including 2 stillbirths (2.4%) and 5 (6.0%) preterm livebirths. Miscarriages, induced abortions, and ectopic pregnancies were observed in 14 (17%), 8 (10%), and 1 (1.2%) pregnancies, respectively. Ovum donation resulted in 5 (6.0%) pregnancies. High maternal age, premature ovarian insufficiency, primary adrenal insufficiency, or hypoparathyroidism did not associate with miscarriages. Women with livebirth had, on average, 4 APECED manifestations (range 0-10); 78% had hypoparathyroidism, and 36% had primary adrenal insufficiency. APECED manifestations remained mostly stable during pregnancy, but in 1 case, development of primary adrenal insufficiency led to adrenal crisis and stillbirth. Birth weights were normal in >80% and apart from 1 neonatal death of a preterm baby, no serious perinatal complications occurred. CONCLUSIONS Outcome of pregnancy in women with APECED was generally favorable. However, APECED warrants careful maternal multidisciplinary follow-up from preconceptual care until puerperium.
Collapse
Affiliation(s)
- Saila Laakso
- Children’s Hospital and Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Correspondence: Saila Laakso, MD, PhD, Children’s Hospital, Stenbäckinkatu 9, FI-00290 Helsinki, Finland.
| | - Elina Holopainen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Corrado Betterle
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua,Italy
| | - Viivi Saari
- Children’s Hospital and Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elinor Vogt
- Department of Clinical Science and K.G. Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Monica M Schmitt
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Karen K Winer
- Eunice Kennedy Shriver National Institutes of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, MD,USA
| | | | - Chiara Sabbadin
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua,Italy
| | - Eystein S Husebye
- Department of Clinical Science and K.G. Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | | | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Outi Mäkitie
- Children’s Hospital and Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| |
Collapse
|
21
|
Pandiyan P, McCormick TS. Regulation of IL-17A-Producing Cells in Skin Inflammatory Disorders. J Invest Dermatol 2021; 142:867-875. [PMID: 34561088 DOI: 10.1016/j.jid.2021.06.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022]
Abstract
This review focuses on the IL-17A family of cytokines produced by T lymphocytes and other immune cells and how they are involved in cutaneous pathogenic responses. It will also discuss cutaneous dysbiosis and FOXP3+ regulatory T cells in the context of inflammatory conditions linked to IL-17 responses in the skin. Specifically, it will review key literature on chronic mucocutaneous candidiasis and psoriasis.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| | - Thomas S McCormick
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
22
|
Berger AH, Bratland E, Sjøgren T, Heimli M, Tyssedal T, Bruserud Ø, Johansson S, Husebye ES, Oftedal BE, Wolff ASB. Transcriptional Changes in Regulatory T Cells From Patients With Autoimmune Polyendocrine Syndrome Type 1 Suggest Functional Impairment of Lipid Metabolism and Gut Homing. Front Immunol 2021; 12:722860. [PMID: 34526996 PMCID: PMC8435668 DOI: 10.3389/fimmu.2021.722860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/12/2021] [Indexed: 01/22/2023] Open
Abstract
Autoimmune polyendocrine syndrome type I (APS-1) is a monogenic model disorder of organ-specific autoimmunity caused by mutations in the Autoimmune regulator (AIRE) gene. AIRE facilitates the expression of organ-specific transcripts in the thymus, which is essential for efficient removal of dangerous self-reacting T cells and for inducing regulatory T cells (Tregs). Although reduced numbers and function of Tregs have been reported in APS-I patients, the impact of AIRE deficiency on gene expression in these cells is unknown. Here, we report for the first time on global transcriptional patterns of isolated Tregs from APS-1 patients compared to healthy subjects. Overall, we found few differences between the groups, although deviant expression was observed for the genes TMEM39B, SKIDA1, TLN2, GPR15, FASN, BCAR1, HLA-DQA1, HLA-DQB1, HLA-DRA, GPSM3 and AKR1C3. Of significant interest, the consistent downregulation of GPR15 may indicate failure of Treg gut homing which could be of relevance for the gastrointestinal manifestations commonly seen in APS-1. Upregulated FASN expression in APS-1 Tregs points to increased metabolic activity suggesting a putative link to faulty Treg function. Functional studies are needed to determine the significance of these findings for the immunopathogenesis of APS-1 and for Treg immunobiology in general.
Collapse
Affiliation(s)
- Amund Holte Berger
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Eirik Bratland
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Thea Sjøgren
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Marte Heimli
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torgeir Tyssedal
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway
| | - Øyvind Bruserud
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Anesthesiology and Intensive Care, Haukeland University Hospital, Bergen, Norway
| | - Stefan Johansson
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Eystein Sverre Husebye
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Bergithe Eikeland Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette Susanne Bøe Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Kristian Gerhard (KG) Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
23
|
Peterson P, Kisand K, Kluger N, Ranki A. Loss of AIRE-Mediated Immune Tolerance and the Skin. J Invest Dermatol 2021; 142:760-767. [PMID: 34535292 DOI: 10.1016/j.jid.2021.04.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023]
Abstract
The core function of the immune response is to distinguish between self and foreign. The multiorgan human autoimmune disease, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED/autoimmune polyendocrine syndrome type 1) is an example of what happens in the body when central immune tolerance goes astray. APECED revealed the existence and function of the autoimmune regulator gene, which has a central role in the development of tolerance. The discovery of autoimmune regulator was the start of a new period in immunology and in understanding the role of central and peripheral tolerance, also very relevant to many skin diseases as we highlight in this review.
Collapse
Affiliation(s)
- Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Nicolas Kluger
- Department of Dermatology, Allergology and Venereology, Clinicum, University of Helsinki, and Inflammation Center, Helsinki University Hospital, Helsinki, Finland
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereology, Clinicum, University of Helsinki, and Inflammation Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
24
|
Zhu F, Willette-Brown J, Zhang J, Ferre EMN, Sun Z, Wu X, Lionakis MS, Hu Y. NLRP3 Inhibition Ameliorates Severe Cutaneous Autoimmune Manifestations in a Mouse Model of Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy-Like Disease. J Invest Dermatol 2021; 141:1404-1415. [PMID: 33188780 PMCID: PMC8110612 DOI: 10.1016/j.jid.2020.10.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/25/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy show diverse endocrine and nonendocrine manifestations initiated by self-reactive T cells because of AIRE mutation-induced defective central tolerance. A large number of American patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy suffer from early-onset cutaneous inflammatory lesions accompanied by an infiltration of T cells and myeloid cells. The role of myeloid cells in this setting remains to be fully investigated. In this study, we characterize the autoinflammatory phenotypes in the skin of both autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy-like kinase-dead Ikkα knockin mice and patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. We found a marked infiltration of autoreactive CD4 T cells, macrophages, and neutrophils; elevated uric acid; and increased NLRP3, a major inflammasome component. Depleting autoreactive CD4 T cells or ablating Ccl2/Cxcr2 genes significantly attenuated the inflammasome activity, inflammation, and skin phenotypes in kinase-dead Ikkα knockin mice. Importantly, treatment with an NLRP3 inhibitor reduced skin phenotypes and decreased infiltration of CD4 T cells, macrophages, and neutrophils. These results suggest that increased myeloid cell infiltration contributes to autoreactive CD4 T cell-mediated skin autoinflammation. Thus, our findings reveal that the combined infiltration of macrophages and neutrophils is required for autoreactive CD4 T cell-mediated skin disease pathogenesis and that the NLRP3-dependent inflammasome is a potential therapeutic target for the cutaneous manifestations of autoimmune diseases.
Collapse
Affiliation(s)
- Feng Zhu
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Jami Willette-Brown
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Jian Zhang
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA; Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Elise M N Ferre
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Zhonghe Sun
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Xiaolin Wu
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Yinling Hu
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA.
| |
Collapse
|
25
|
Zhan F, Cao L. Late-onset autoimmune polyendocrine syndrome type 1: a case report and literature review. Immunol Res 2021; 69:139-144. [PMID: 33599910 PMCID: PMC7889704 DOI: 10.1007/s12026-021-09180-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/07/2021] [Indexed: 01/07/2023]
Abstract
Autoimmune polyendocrine syndrome type 1 (APS-1), also referred to as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), a rare monogenic disorder, is classically characterized by a triad of chronic mucocutaneous candidiasis, hypoparathyroidism, and primary adrenal insufficiency. The identified causative gene is autoimmune regulator (AIRE), which encodes a critical transcription factor and is essential for self-tolerance. Here, we describe a late-onset Chinese case who presented with symptoms of persistent tetany due to hypocalcemia. Extensive clinical evaluations revealed that the patient manifested beyond the classic triad of the disease, and next-generation sequencing identified a known homozygous AIRE mutation (p.R139X). APS-1 is a rare inherited immunodeficiency disease with high clinical and genetic heterogeneity. By retrospectively analyzing the disease, we comprehensively reviewed the phenotypic features, summarized the genotype spectrum, and discussed the possible immunological mechanisms of the disease to enhance earlier recognition and implement targeted preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Feixia Zhan
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, 600 Yi Shan Road, Shanghai, 200233 China
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai, 200003 China
| | - Li Cao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, 600 Yi Shan Road, Shanghai, 200233 China
| |
Collapse
|
26
|
Fierabracci A, Arena A, Toto F, Gallo N, Puel A, Migaud M, Kumar M, Chengappa KG, Gulati R, Negi VS, Betterle C. Autoimmune polyendocrine syndrome type 1 (APECED) in the Indian population: case report and review of a series of 45 patients. J Endocrinol Invest 2021; 44:661-677. [PMID: 32767280 DOI: 10.1007/s40618-020-01376-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Autoimmune polyendocrinopathy-candidiasis-ectodermal-dystrophy (APECED) or autoimmune polyglandular syndrome type 1 (APS-1) is a rare autosomal recessive genetic disease due to mutations in the AIRE (AutoImmune REgulator) gene. The clinical diagnosis is classically based on the presence of at least two of the three main components: chronic mucocutaneous candidiasis, hypoparathyroidism and primary adrenal insufficiency. Patients often suffer from other endocrine or non-endocrine autoimmune conditions throughout life. APECED etiopathogenesis is mediated by T lymphocytes. Autoantibodies against proteins of the affected organs are found in the serum of APECED patients as well as neutralizing antibodies against cytokines. We report here the clinical and genetic characteristics of 45 Indian APECED patients in comparison to Finnish, Sardinian, Turkish and North/South American cohorts from their published results. We also report a new case of APECED of Indian origin, a 2-year old child suffering from chronic mucocutaneous candidiasis since the age of 8 months, with confirmatory AIRE homozygous mutation c.274C > T (p.R92W). CONCLUSION With the inherent limitations of a retrospective study, analysis of Indian APECED patients suggested that compared to classic criteria, application of Ferre/Lionakis criteria validated in North/South American patients could help in earlier diagnosis in 3 of 8 (37.5%) patients for whom adequate information for evaluation was available.
Collapse
Affiliation(s)
- A Fierabracci
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - A Arena
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - F Toto
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - N Gallo
- Laboratory Medicine, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - A Puel
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR 1163, Imagine Institute, University of Paris, Paris, France
| | - M Migaud
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR 1163, Imagine Institute, University of Paris, Paris, France
| | - M Kumar
- Department of Clinical Immunology, JIPMER, Pondicherry, India
| | - K G Chengappa
- Department of Clinical Immunology, JIPMER, Pondicherry, India
| | - R Gulati
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - V S Negi
- Department of Clinical Immunology, JIPMER, Pondicherry, India
| | - C Betterle
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| |
Collapse
|
27
|
Bacchetta R, Weinberg K. Thymic origins of autoimmunity-lessons from inborn errors of immunity. Semin Immunopathol 2021; 43:65-83. [PMID: 33532929 PMCID: PMC7925499 DOI: 10.1007/s00281-020-00835-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022]
Abstract
During their intrathymic development, nascent T cells are empowered to protect against pathogens and to be operative for a life-long acceptance of self. While autoreactive effector T (Teff) cell progenitors are eliminated by clonal deletion, the intrathymic mechanisms by which thymic regulatory T cell (tTreg) progenitors maintain specificity for self-antigens but escape deletion to exert their regulatory functions are less well understood. Both tTreg and Teff development and selection result from finely coordinated interactions between their clonotypic T cell receptors (TCR) and peptide/MHC complexes expressed by antigen-presenting cells, such as thymic epithelial cells and thymic dendritic cells. tTreg function is dependent on expression of the FOXP3 transcription factor, and induction of FOXP3 gene expression by tTreg occurs during their thymic development, particularly within the thymic medulla. While initial expression of FOXP3 is downstream of TCR activation, constitutive expression is fixed by interactions with various transcription factors that are regulated by other extracellular signals like TCR and cytokines, leading to epigenetic modification of the FOXP3 gene. Most of the understanding of the molecular events underlying tTreg generation is based on studies of murine models, whereas gaining similar insight in the human system has been very challenging. In this review, we will elucidate how inborn errors of immunity illuminate the critical non-redundant roles of certain molecules during tTreg development, shedding light on how their abnormal development and function cause well-defined diseases that manifest with autoimmunity alone or are associated with states of immune deficiency and autoinflammation.
Collapse
Affiliation(s)
- Rosa Bacchetta
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Lokey Stem Cell Research Building 265 Campus Drive, West Stanford, CA, 94305, USA.
- Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Kenneth Weinberg
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Lokey Stem Cell Research Building 265 Campus Drive, West Stanford, CA, 94305, USA
- Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
28
|
Gong J, Zeng Q, Yu D, Duan YG. T Lymphocytes and Testicular Immunity: A New Insight into Immune Regulation in Testes. Int J Mol Sci 2020; 22:ijms22010057. [PMID: 33374605 PMCID: PMC7793097 DOI: 10.3390/ijms22010057] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
The immune privilege of the testes is necessary to prevent immune attacks to gamete-specific antigens and paternal major histocompatibility complex (MHC) antigens, allowing for normal spermatogenesis. However, infection and inflammation of the male genital tract can break the immune tolerance and represent a significant cause of male infertility. Different T cell subsets have been identified in mammalian testes, which may be involved in the maintenance of immune tolerance and pathogenic immune responses in testicular infection and inflammation. We reviewed the evidence in the published literature on different T subtypes (regulatory T cells, helper T cells, cytotoxic T cells, γδ T cells, and natural killer T cells) in human and animal testes that support their regulatory roles in infertility and the orchitis pathology. While many in vitro studies have indicated the regulation potential of functional T cell subsets and their possible interaction with Sertoli cells, Leydig cells, and spermatogenesis, both under physiological and pathological processes, there have been no in situ studies to date. Nevertheless, the normal distribution and function of T cell subsets are essential for the immune privilege of the testes and intact spermatogenesis, and T cell-mediated immune response drives testicular inflammation. The distinct function of different T cell subsets in testicular homeostasis and the orchitis pathology suggests a considerable potential of targeting specific T cell subsets for therapies targeting chronic orchitis and immune infertility.
Collapse
Affiliation(s)
- Jialei Gong
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Qunxiong Zeng
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
29
|
Borchers J, Pukkala E, Mäkitie O, Laakso S. Patients With APECED Have Increased Early Mortality Due to Endocrine Causes, Malignancies and infections. J Clin Endocrinol Metab 2020; 105:5809346. [PMID: 32185376 PMCID: PMC7150614 DOI: 10.1210/clinem/dgaa140] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/16/2020] [Indexed: 01/12/2023]
Abstract
CONTEXT Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is an autoimmune endocrinopathy with severe and unpredictable course. The impact of APECED on mortality has not been determined. OBJECTIVE To assess overall and cause-specific mortality of patients with APECED. DESIGN AND SETTING A follow-up study of Finnish patients with APECED from 1971 to 2018. Causes and dates of death were collected from Finnish registries. PATIENTS Ninety-one patients with APECED. MAIN OUTCOME MEASURE Overall and cause-specific standardized mortality ratios (SMRs) determined by comparing the observed numbers of death and those expected on the basis of respective population death rates in Finland. RESULTS The overall disease mortality was significantly increased (29 deaths, SMR 11; 95% confidence interval [CI] 7.2-16; P < 0.001). The relative risk (SMR) was highest in the youngest age groups but the absolute excess risk was similar (about 10 per 10 000 person-years) in all age categories. The highest SMRs were seen for endocrine and metabolic diseases (SMR 570; 95% CI, 270-1000; P < 0.001) and for oral and esophageal malignancies (SMR 170; 95% CI, 68-360; P < 0.001). Mortality was also increased for infections, diseases of digestive system, alcohol-related deaths, and for accidents. Due to the small number of cases we were unable to evaluate whether mortality was affected by disease severity. CONCLUSIONS Patients with APECED have significantly increased mortality in all age groups. Highest SMRs are found for causes that are directly related to APECED but also for infections. Increased alcohol- and accident-related deaths may be influenced by psychosocial factors.
Collapse
Affiliation(s)
- Joonatan Borchers
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eero Pukkala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
- Finnish Cancer Registry – Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - Outi Mäkitie
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Saila Laakso
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Correspondence and Reprint Requests: Saila Laakso, MD, PhD, PO. Box 347, 00029 HUS, Finland. E-mail:
| |
Collapse
|
30
|
Sakaguchi S, Mikami N, Wing JB, Tanaka A, Ichiyama K, Ohkura N. Regulatory T Cells and Human Disease. Annu Rev Immunol 2020; 38:541-566. [PMID: 32017635 DOI: 10.1146/annurev-immunol-042718-041717] [Citation(s) in RCA: 693] [Impact Index Per Article: 138.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Naturally occurring CD4+ regulatory T cells (Tregs), which specifically express the transcription factor FoxP3 in the nucleus and CD25 and CTLA-4 on the cell surface, are a functionally distinct T cell subpopulation actively engaged in the maintenance of immunological self-tolerance and homeostasis. Recent studies have facilitated our understanding of the cellular and molecular basis of their generation, function, phenotypic and functional stability, and adaptability. It is under investigation in humans how functional or numerical Treg anomalies, whether genetically determined or environmentally induced, contribute to immunological diseases such as autoimmune diseases. Also being addressed is how Tregs can be targeted to control physiological and pathological immune responses, for example, by depleting them to enhance tumor immunity or by expanding them to treat immunological diseases. This review discusses our current understanding of Treg immunobiology in normal and disease states, with a perspective on the realization of Treg-targeting therapies in the clinic.
Collapse
Affiliation(s)
- Shimon Sakaguchi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; .,Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Norihisa Mikami
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - James B Wing
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - Atsushi Tanaka
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - Kenji Ichiyama
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - Naganari Ohkura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| |
Collapse
|
31
|
Laakso S, Borchers J, Toiviainen-Salo S, Pekkinen M, Mäkitie O. Severe Phenotype of APECED (APS1) Increases Risk for Structural Bone Alterations. Front Endocrinol (Lausanne) 2020; 11:109. [PMID: 32210917 PMCID: PMC7067693 DOI: 10.3389/fendo.2020.00109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 02/19/2020] [Indexed: 12/31/2022] Open
Abstract
Objective: Immunological abnormalities, the resulting endocrinopathies and their treatments may impact bone health in patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED, APS1). The aim of the present study was to describe skeletal characteristics in patients with APECED and the prevalence and risk factors of compromised bone health. Patients and methods: We performed a cross-sectional study on 44 patients (27 females) with APECED and 82 age-, gender- and ethnicity-matched control subjects (54 females). We determined the prevalence of osteoporosis by dual-energy X-ray absorptiometry and skeletal characteristics by peripheral quantitative computed tomography at radius and tibia. Results: Patients were examined at the median age of 37.8 years (range, 7.0-70.1). Dual-energy X-ray absorptiometry indicated osteoporosis in four adult patients (9%); radiographs showed vertebral fractures in three patients. The prevalence of multiple non-spinal fractures was higher in patients than in controls. On peripheral quantitative computed tomography, bone characteristics at distal and proximal radius did not differ between the groups. At distal tibia, patients had lower total (p = 0.009) and trabecular (p = 0.033) volumetric bone mineral density. At the proximal tibia, patients had lower cortical thickness (p < 0.001) than controls. Severity of APECED phenotype influenced both radial and tibial characteristics: cortical thickness and total and trabecular volumetric bone mineral density were lower in patients with ≥7 disease manifestations as compared with more mildly affected patients, whose values were similar to controls. Conclusions: APECED associated with bone structural alterations, especially in patients with a high number of disease manifestations. This may increase the risk of fractures with aging, but symptomatic osteoporosis was rare.
Collapse
MESH Headings
- Absorptiometry, Photon
- Adolescent
- Adult
- Aged
- Bone Density
- Bone Diseases, Metabolic/diagnosis
- Bone Diseases, Metabolic/epidemiology
- Bone Diseases, Metabolic/etiology
- Child
- Cross-Sectional Studies
- Female
- Fractures, Bone/diagnosis
- Fractures, Bone/epidemiology
- Fractures, Bone/etiology
- Humans
- Male
- Middle Aged
- Osteoporosis/diagnosis
- Osteoporosis/epidemiology
- Osteoporosis/etiology
- Phenotype
- Polyendocrinopathies, Autoimmune/complications
- Polyendocrinopathies, Autoimmune/epidemiology
- Polyendocrinopathies, Autoimmune/pathology
- Prevalence
- Risk Factors
- Severity of Illness Index
- Young Adult
Collapse
Affiliation(s)
- Saila Laakso
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- *Correspondence: Saila Laakso
| | - Joonatan Borchers
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sanna Toiviainen-Salo
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pediatric Radiology, Medical Imaging Center, Helsinki University Hospital, Helsinki, Finland
| | - Minna Pekkinen
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Outi Mäkitie
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
32
|
Zhang P, Liu RT, Du T, Yang CL, Liu YD, Ge MR, Zhang M, Li XL, Li H, Dou YC, Duan RS. Exosomes derived from statin-modified bone marrow dendritic cells increase thymus-derived natural regulatory T cells in experimental autoimmune myasthenia gravis. J Neuroinflammation 2019; 16:202. [PMID: 31679515 PMCID: PMC6825716 DOI: 10.1186/s12974-019-1587-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The thymus plays an essential role in the pathogenesis of myasthenia gravis (MG). In patients with MG, natural regulatory T cells (nTreg), a subpopulation of T cells that maintain tolerance to self-antigens, are severely impaired in the thymuses. In our previous study, upregulated nTreg cells were observed in the thymuses of rats in experimental autoimmune myasthenia gravis after treatment with exosomes derived from statin-modified dendritic cells (statin-Dex). METHODS We evaluated the effects of exosomes on surface co-stimulation markers and Aire expression of different kinds of thymic stromal cells, including cTEC, mTEC, and tDCs, in EAMG rats. The isolated exosomes were examined by western blot and DLS. Immunofluorescence was used to track the exosomes in the thymus. Flow cytometry and western blot were used to analyze the expression of co-stimulatory molecules and Aire in vivo and in vitro. RESULTS We confirmed the effects of statin-Dex in inducing Foxp3+ nTreg cells and found that both statin-Dex and DMSO-Dex could upregulate CD40 but only statin-Dex increased Aire expression in thymic stromal cells in vivo. Furthermore, we found that the role of statin-Dex and DMSO-Dex in the induction of Foxp3+ nTreg cells was dependent on epithelial cells in vitro. CONCLUSIONS We demonstrated that statin-Dex increased expression of Aire in the thymus, which may further promote the Foxp3 expression in the thymus. These findings may provide a new strategy for the treatment of myasthenia gravis.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Ru-Tao Liu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Tong Du
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Chun-Lin Yang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Yu-Dong Liu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Meng-Ru Ge
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Min Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Xiao-Li Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Heng Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
| | - Ying-Chun Dou
- College of Basic Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355 People’s Republic of China
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014 People’s Republic of China
- Department of Neurology, the First Affiliated Hospital of Shandong First Medical University, Jinan, 250014 People’s Republic of China
| |
Collapse
|
33
|
Owen DL, Sjaastad LE, Farrar MA. Regulatory T Cell Development in the Thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:2031-2041. [PMID: 31591259 PMCID: PMC6910132 DOI: 10.4049/jimmunol.1900662] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022]
Abstract
Development of a comprehensive regulatory T (Treg) cell compartment in the thymus is required to maintain immune homeostasis and prevent autoimmunity. In this study, we review cellular and molecular determinants of Treg cell development in the thymus. We focus on the evidence for a self-antigen-focused Treg cell repertoire as well as the APCs responsible for presenting self-antigens to developing thymocytes. We also cover the contribution of different cytokines to thymic Treg development and the cellular populations that produce these cytokines. Finally, we update the originally proposed "two-step" model of thymic Treg differentiation by incorporating new evidence demonstrating that Treg cells develop from two Treg progenitor populations and discuss the functional importance of Treg cells generated via either progenitor pathway.
Collapse
Affiliation(s)
- David L Owen
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Louisa E Sjaastad
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Michael A Farrar
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
34
|
Abstract
With the advent of the concept of dominant tolerance and the subsequent discovery of CD4+ regulatory T cells expressing the transcription factor FOXP3 (Tregs), almost all productive as well as nonproductive immune responses can be compartmentalized to a binary of immune effector T cells and immune regulatory Treg populations. A beneficial immune response warrants the timely regulation by Tregs, whereas a nonproductive immune response indicates insufficient effector functions or an outright failure of tolerance. There are ample reports supporting role of Tregs in suppressing spontaneous auto-immune diseases as well as promoting immune evasion by cancers. To top up their importance, several non-immune functions like tissue homeostasis and regeneration are also being attributed to Tregs. Hence, after being in the center stage of basic and translational immunological research, Tregs are making the next jump towards clinical studies. Therefore, newer small molecules, biologics as well as adoptive cell therapy (ACT) approaches are being tested to augment or undermine Treg responses in the context of autoimmunity and cancer. In this brief review, we present the strategies to modulate Tregs towards a favorable clinical outcome.
Collapse
Affiliation(s)
- Amit Sharma
- Academy of Immunology and Microbiology, Institute for Basic Science (IBS) , Pohang , Republic of Korea.,Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology (POSTECH) , Pohang , Republic of Korea
| | - Dipayan Rudra
- Academy of Immunology and Microbiology, Institute for Basic Science (IBS) , Pohang , Republic of Korea.,Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology (POSTECH) , Pohang , Republic of Korea
| |
Collapse
|
35
|
Su MA, Anderson MS. Pulling RANK on Cancer: Blocking Aire-Mediated Central Tolerance to Enhance Immunotherapy. Cancer Immunol Res 2019; 7:854-859. [PMID: 31160305 PMCID: PMC6550349 DOI: 10.1158/2326-6066.cir-18-0912] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A major breakthrough in cancer treatment occurred with the development of strategies that overcome T-cell tolerance toward tumor cells. These approaches enhance antitumor immunity by overcoming mechanisms that are normally in place to prevent autoimmunity but simultaneously prevent rejection of tumor cells. Although tolerance mechanisms that restrict antitumor immunity take place both in the thymus and periphery, only immunotherapies that target peripheral tolerance mechanisms occurring outside of the thymus are currently available. We review here recent gains in our understanding of how thymic tolerance mediated by the autoimmune regulator (Aire) impedes antitumor immunity. It is now clear that transient depletion of Aire-expressing cells in the thymus can be achieved with RANKL blockade. Finally, we discuss key findings that support the repurposing of anti-RANKL as a cancer immunotherapy with a unique mechanism of action.
Collapse
Affiliation(s)
- Maureen A Su
- Microbiology, Immunology, and Medical Genetics and Pediatrics, University of California, Los Angeles, Los Angeles, California.
| | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
36
|
Sng J, Ayoglu B, Chen JW, Schickel JN, Ferre EMN, Glauzy S, Romberg N, Hoenig M, Cunningham-Rundles C, Utz PJ, Lionakis MS, Meffre E. AIRE expression controls the peripheral selection of autoreactive B cells. Sci Immunol 2019; 4:eaav6778. [PMID: 30979797 PMCID: PMC7257641 DOI: 10.1126/sciimmunol.aav6778] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/04/2019] [Indexed: 12/13/2022]
Abstract
Autoimmune regulator (AIRE) mutations result in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) syndrome characterized by defective central T cell tolerance and the production of many autoantibodies targeting tissue-specific antigens and cytokines. By studying CD3- and AIRE-deficient patients, we found that lack of either T cells or AIRE function resulted in the peripheral accumulation of autoreactive mature naïve B cells. Proteomic arrays and Biacore affinity measurements revealed that unmutated antibodies expressed by these autoreactive naïve B cells recognized soluble molecules and cytokines including insulin, IL-17A, and IL-17F, which are AIRE-dependent thymic peripheral tissue antigens targeted by autoimmune responses in APECED. AIRE-deficient patients also displayed decreased frequencies of regulatory T cells (Tregs) that lacked common TCRβ clones found instead in their conventional T cell compartment, thereby suggesting holes in the Treg TCR repertoire of these patients. Hence, AIRE-mediated T cell/Treg selection normally prevents the expansion of autoreactive naïve B cells recognizing peripheral self-antigens.
Collapse
Affiliation(s)
- Joel Sng
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Burcu Ayoglu
- School of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA 94305, USA
| | - Jeff W Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Jean-Nicolas Schickel
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Elise M N Ferre
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Salomé Glauzy
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Neil Romberg
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Manfred Hoenig
- Department of Pediatrics, University Medical Centre Ulm, Ulm, Germany
| | - Charlotte Cunningham-Rundles
- Division of Allergy and Immunology, Department of Medicine, Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
| | - Paul J Utz
- School of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA 94305, USA
- Institute for Immunity, Transplantation, and Infection (ITI), Stanford University, Stanford, CA 94305, USA
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
37
|
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: 102] [Impact Index Per Article: 17.0] [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
|
38
|
Jacobo P. The role of regulatory T Cells in autoimmune orchitis. Andrologia 2018; 50:e13092. [DOI: 10.1111/and.13092] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/06/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Patricia Jacobo
- Departmental and Institutional Affiliation, Instituto de Investigaciones Biomédicas, UBA-CONICET, Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| |
Collapse
|
39
|
Weiler FG, Peterson P, Costa-Carvalho BT, de Barros Dorna M, Correia-Deur JE, Sader SL, Espíndola-Antunes D, Guerra-Junior G, Dias-da-Silva MR, Lazaretti-Castro M. The heterogeneity of autoimmune polyendocrine syndrome type 1: Clinical features, new mutations and cytokine autoantibodies in a Brazilian cohort from tertiary care centers. Clin Immunol 2018; 197:231-238. [PMID: 30287219 DOI: 10.1016/j.clim.2018.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 09/16/2018] [Accepted: 09/26/2018] [Indexed: 12/31/2022]
Abstract
Autoimmune polyendocrine syndrome type 1 (APS1) is characterized by multiorgan autoimmunity. We aim at characterizing a multi-center Brazilian cohort of APS1 patients by clinical evaluation, searching mutation in the AIRE gene, measuring serum autoantibodies, and investigating correlations between findings. We recruited patients based on the clinical criteria and tested them for AIRE mutations, antibodies against interferon type I and interleukins 17A, 17F and 22. We identified 12 unrelated families (13 patients) with typical signs of APS1 in the proband, and the screening of relatives recognized an asymptomatic child. Candidiasis was present in all cases, and 19 other manifestations were observed. All patients carried one of 10 different mutations in AIRE, being 3 new ones, and were positive for anti-interferon type I serum antibody. Anti-interleukin-17A levels inversely correlated with the number of manifestations in each patient. This negative correlation may suggest a protective effect of anti-interleukin-17A with a potential therapeutic application.
Collapse
Affiliation(s)
- Fernanda Guimarães Weiler
- Division of Endocrinology, Department of Medicine, Universidade Federal de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | | | - Mayra de Barros Dorna
- Allergy and immunology unit, Department of Pediatrics, Universidade de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | | | - Soraya Lopes Sader
- Department of Pediatrics, Ribeirao Preto Medical School, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Gil Guerra-Junior
- Department of Pediatrics, Universidade Estadual de Campinas, Campinas, Sao Paulo, Brazil
| | - Magnus Régios Dias-da-Silva
- Division of Endocrinology, Department of Medicine, Universidade Federal de Sao Paulo, Sao Paulo, Sao Paulo, Brazil.
| | - Marise Lazaretti-Castro
- Division of Endocrinology, Department of Medicine, Universidade Federal de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
40
|
Fierabracci A, Pellegrino M, Frasca F, Kilic SS, Betterle C. APECED in Turkey: A case report and insights on genetic and phenotypic variability. Clin Immunol 2018; 194:60-66. [PMID: 30018023 DOI: 10.1016/j.clim.2018.06.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 06/27/2018] [Accepted: 06/29/2018] [Indexed: 01/06/2023]
Abstract
APECED is a rare monogenic recessive disorder caused by mutations in the AIRE gene. In this manuscript, we report a male Turkish patient with APECED syndrome who presented with chronic mucocutaneous candidiasis associated with other autoimmune manifestations developed over the years. The presence of the homozygous R257X mutation of the AIRE gene confirmed the diagnosis of APECED syndrome. We further performed literature review in 23 published Turkish APECED patients and noted that Finnish major mutation R257X is common in Turks. In particular, we assessed retrospectively how often the Ferre/Lionakis criteria would have resulted in earlier diagnosis in Finns, Sardinians and Turks in respect to the classic criteria. Since an earlier diagnosis could have been possible in 18.8% of Turkish, in 23.8% of Sardinian and 38.55% of Finnish patients we reviewed from literature, Ferre/Lionakis criteria could indeed allow in future earlier initiation of immunomodulatory treatments, if found effective in future studies.
Collapse
Affiliation(s)
- Alessandra Fierabracci
- Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Viale S. Paolo 15, 00146 Rome, Italy.
| | - Marsha Pellegrino
- Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Viale S. Paolo 15, 00146 Rome, Italy.
| | - Federica Frasca
- Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Viale S. Paolo 15, 00146 Rome, Italy.
| | - Sara Sebnem Kilic
- Pediatric Immunology -Rheumatology, Uludag University, Medical Faculty Department of Pediatrics, Gorukle-Bursa 16059, Turkey.
| | - Corrado Betterle
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Via Ospedale Civile 105, 35128 Padua, Italy.
| |
Collapse
|
41
|
Pellegrino M, Bellacchio E, Dhamo R, Frasca F, Betterle C, Fierabracci A. A Novel Homozygous Mutation of the AIRE Gene in an APECED Patient From Pakistan: Case Report and Review of the Literature. Front Immunol 2018; 9:1835. [PMID: 30150985 PMCID: PMC6099424 DOI: 10.3389/fimmu.2018.01835] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/25/2018] [Indexed: 11/13/2022] Open
Abstract
Autoimmune-poly-endocrinopathy-candidiasis-ectodermal-dystrophy syndrome (APECED) is a rare monogenic recessive disorder caused by mutations in the autoimmune regulator (AIRE) gene. Criteria for the diagnosis of APECED are the presence of two of the following disorders: chronic mucocutaneous candidiasis (CMC), chronic hypoparathyroidism (CHP), and Addison's disease. APECED develops at high incidence in Finns, Sardinians, and Iranian Jews and presents with a wide range of clinical phenotypes and genotypes. In this manuscript, we report the clinical, endocrinological, and molecular features of a 16-year-old female patient from Pakistan living in Italy and presenting the major APECED clinical manifestations CMC, CHP, and primary adrenal insufficiency. Premature ovarian failure, chronic bronchopneumopathy, vitiligo, Hashimoto's thyroiditis emerged as associated diseases. In our patient, AIRE gene screening revealed the novel c.396G>C (p.Arg132Ser; p.R132S) mutation in homozygosity thus confirming APECED diagnosis. This is the first reported mutation within the nuclear localization signal (NLS) that is associated with APECED. The NLS mutation affects the nuclear import of classical transcription factors through nuclear pore by recognition of nuclear import receptors, the importin α molecules. By displaying crystal structures of the peptide containing the KRK basic residue cluster bound to α importins, we show that p.R132S replacement in 131-KRK-133 does not reproduce these interactions. Thus, we propose that the novel mutation exerts its pathogenetic effect by impairing the nuclear import of the Aire protein. The present case report is added to a limited series of Pakistani APECED patients who we reviewed from the scientific literature, mostly diagnosed on clinical findings.
Collapse
Affiliation(s)
- Marsha Pellegrino
- Infectivology and Clinical Trials Research Division, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Emanuele Bellacchio
- Molecular Genetics and Functional Genomics, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, Rome, Italy
| | | | - Federica Frasca
- Infectivology and Clinical Trials Research Division, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Corrado Betterle
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Research Division, Bambino Gesù Children’s Hospital, Rome, Italy
| |
Collapse
|
42
|
Azizi G, Yazdani R, Rae W, Abolhassani H, Rojas M, Aghamohammadi A, Anaya JM. Monogenic polyautoimmunity in primary immunodeficiency diseases. Autoimmun Rev 2018; 17:1028-1039. [PMID: 30107266 DOI: 10.1016/j.autrev.2018.05.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 02/08/2023]
Abstract
Primary immunodeficiency diseases (PIDs) consist of a large group of genetic disorders that affect distinct components of the immune system. PID patients are susceptible to infection and non-infectious complications, particularly autoimmunity. A specific group of monogenic PIDs are due to mutations in genes that are critical for the regulation of immunological tolerance and immune responses. This group of monogenic PIDs is at high risk of developing polyautoimmunity (i.e., the presence of more than one autoimmune disease in a single patient) because of their impaired immunity. In this review, we discuss the mechanisms of autoimmunity in PIDs and the characteristics of polyautoimmunity in the following PIDs: IPEX; monogenic IPEX-like syndrome; LRBA deficiency; CTLA4 deficiency; APECED; ALPS; and PKCδ deficiency.
Collapse
Affiliation(s)
- Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Wiliam Rae
- Department of Immunology, MP8, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, Hampshire SO16 6YD, UK
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia.
| |
Collapse
|
43
|
Pohar J, Simon Q, Fillatreau S. Antigen-Specificity in the Thymic Development and Peripheral Activity of CD4 +FOXP3 + T Regulatory Cells. Front Immunol 2018; 9:1701. [PMID: 30083162 PMCID: PMC6064734 DOI: 10.3389/fimmu.2018.01701] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/10/2018] [Indexed: 01/12/2023] Open
Abstract
CD4+Foxp3+ T regulatory cells (Treg) are essential for the life of the organism, in particular because they protect the host against its own autoaggressive CD4+Foxp3- T lymphocytes (Tconv). Treg distinctively suppress autoaggressive immunity while permitting efficient defense against infectious diseases. This split effect indicates that Treg activity is controlled in an antigen-specific manner. This specificity is achieved first by the formation of the Treg repertoire during their development, and second by their activation in the periphery. This review presents novel information on the antigen-specificity of Treg development in the thymus, and Treg function in the periphery. These aspects have so far remained imprecisely understood due to the lack of knowledge of the actual antigens recognized by Treg during the different steps of their life, so that most previous studies have been performed using artificial antigens. However, recent studies identified some antigens mediating the positive selection of autoreactive Treg in the thymus, and the function of Treg in the periphery in autoimmune and allergic disorders. These investigations emphasized the remarkable specificity of Treg development and function. Indeed, the development of autoreactive Treg in the thymus was found to be mediated by single autoantigens, so that the absence of one antigen led to a dramatic loss of Treg reacting toward that antigen. The specificity of Treg development is important because the constitution of the Treg repertoire, and especially the presence of holes in this repertoire, was found to crucially influence human immunopathology. Indeed, it was found that the development of human immunopathology was permitted by the lack of Treg against the antigens driving the autoimmune or allergic T cell responses rather than by the impairment of Treg activation or function. The specificity of Treg suppression in the periphery is therefore intimately associated with the mechanisms shaping the formation of the Treg repertoire during their development. This novel information refines significantly our understanding of the antigen-specificity of Treg protective function, which is required to envision how these cells distinctively regulate unwanted immune responses as well as for the development of appropriate approaches to optimally harness them therapeutically in autoimmune, malignant, and infectious diseases.
Collapse
Affiliation(s)
- Jelka Pohar
- Institut Necker-Enfants Malades, INSERM U1151-CNRS UMR 8253, Paris, France
| | - Quentin Simon
- Institut Necker-Enfants Malades, INSERM U1151-CNRS UMR 8253, Paris, France
| | - Simon Fillatreau
- Institut Necker-Enfants Malades, INSERM U1151-CNRS UMR 8253, Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,AP-HP, Hôpital Necker Enfants Malades, Paris, France
| |
Collapse
|
44
|
Conteduca G, Indiveri F, Filaci G, Negrini S. Beyond APECED: An update on the role of the autoimmune regulator gene (AIRE) in physiology and disease. Autoimmun Rev 2018; 17:325-330. [PMID: 29427825 DOI: 10.1016/j.autrev.2017.10.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/24/2017] [Indexed: 12/15/2022]
Abstract
The autoimmune regulator gene (AIRE) is a transcription factor expressed both in the thymus, by medullary thymic epithelial cells, and in secondary lymphoid organs. AIRE controls the local transcription of organ- specific proteins typically expressed in peripheral tissues, thus allowing the negative selection of self- reactive T cells. The crucial role played by AIRE in central immune tolerance emerged in the studies on the pathogenesis of Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy, a rare inherited polyendocrine/autoimmune disease. Thereafter, several studies found evidences indicating that AIRE impairment might be pathogenically involved in several autoimmune diseases and in tumorigenesis. In this review, we focus on recent advances relative to AIRE's effect on T cell development in physiology and disease. In particular, we address the following issues: 1) AIRE function and mTECs biology, 2) the impact of AIRE gene mutations in autoimmune diseases, and 3) the role of AIRE gene in anti-tumor immune response.
Collapse
Affiliation(s)
- Giuseppina Conteduca
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, Laboratory of Hematology, University of Liège, Liège, Belgium
| | - Francesco Indiveri
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Internal Medicine, Clinical Immunology Unit, University of Genoa, Genoa, Italy
| | - Gilberto Filaci
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Internal Medicine, Clinical Immunology Unit, University of Genoa, Genoa, Italy.
| | - Simone Negrini
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Internal Medicine, Clinical Immunology Unit, University of Genoa, Genoa, Italy
| |
Collapse
|
45
|
Berrih-Aknin S, Panse RL, Dragin N. AIRE: a missing link to explain female susceptibility to autoimmune diseases. Ann N Y Acad Sci 2018; 1412:21-32. [PMID: 29291257 DOI: 10.1111/nyas.13529] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 12/13/2022]
Abstract
Women are more susceptible to autoimmune diseases than men. Autoimmunity results from tolerance breakdown toward self-components. Recently, three transcription modulators were identified in medullary thymic epithelial cells that orchestrate immune central tolerance processes: the autoimmune regulator (AIRE), FEZ family zinc finger 2 (FEZF2 or FEZ1), and PR domain zinc finger protein 1 (PRDM1). Interestingly, these three transcription modulators regulate nonredundant tissue-specific antigen subsets and thus cover broad antigen diversity. Recent data from different groups demonstrated that sex hormones (estrogen and testosterone) are involved in the regulation of thymic AIRE expression in humans and mice through direct transcriptional modulation and epigenetic changes. As a consequence, AIRE displays gender-biased thymic expression, with females showing a lower expression compared with males, a finding that could explain the female susceptibility to autoimmune diseases. So far, FEZF2 has not been related to an increased gender bias in autoimmune disease. PRDM1 expression has not been shown to display gender-differential thymic expression, but its expression level and its gene polymorphisms are associated with female-dependent autoimmune disease risk. Altogether, various studies have demonstrated that increased female susceptibility to autoimmune diseases is in part a consequence of hormone-driven reduced thymic AIRE expression.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Rozen Le Panse
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Nadine Dragin
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
- Inovarion, Paris, France
| |
Collapse
|
46
|
Koivula TT, Laakso SM, Niemi HJ, Kekäläinen E, Laine P, Paulin L, Auvinen P, Arstila TP. Clonal Analysis of Regulatory T Cell Defect in Patients with Autoimmune Polyendocrine Syndrome Type 1 Suggests Intrathymic Impairment. Scand J Immunol 2017; 86:221-228. [PMID: 28736829 DOI: 10.1111/sji.12586] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 07/14/2017] [Indexed: 01/28/2023]
Abstract
Mutations in the autoimmune regulator gene disrupt thymic T cell development and negative selection, leading to the recessively inherited polyendocrine autoimmune disease autoimmune polyendocrine syndrome type 1 (APS-1). The patients also have a functional defect in the FOXP3+ regulatory T cell population, but its origin is unclear. Here, we have used T cell receptor sequencing to analyse the clonal relationship of major CD4+ T cell subsets in three patients and three healthy controls. The naive regulatory T cells showed little overlap with helper T cell subsets, supporting divergence in the thymus. The activated/memory regulatory T cell subset displayed more sharing with helper T cells, but was mainly recruited from the naive regulatory T cell population. These clonal patterns were very similar in both patients and controls. However, naive regulatory T cells isolated from the patients had a significantly longer T cell receptor complementarity-determining region 3 than any other population, suggesting failure of thymic selection. These data indicate that the peripheral differentiation of regulatory T cells in APS-1 patients is not different from that in healthy controls. Rather, the patients' naive regulatory T cells may have an intrinsic defect imprinted already in the thymus.
Collapse
Affiliation(s)
- T-T Koivula
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| | - S M Laakso
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| | - H J Niemi
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| | - E Kekäläinen
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
- HUSLAB, Division of Clinical Microbiology, Helsinki University Hospital, HUS, Helsinki, Finland
| | - P Laine
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - L Paulin
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - P Auvinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - T P Arstila
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
47
|
Systemic Activation of NRF2 Alleviates Lethal Autoimmune Inflammation in Scurfy Mice. Mol Cell Biol 2017; 37:MCB.00063-17. [PMID: 28507037 DOI: 10.1128/mcb.00063-17] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 05/11/2017] [Indexed: 12/20/2022] Open
Abstract
The transcription factor NRF2 (nuclear factor [erythroid-derived 2]-like 2) plays crucial roles in the defense mechanisms against oxidative stress and mediates anti-inflammatory actions under various pathological conditions. Recent studies showed that the dysfunction of regulatory T cells (Tregs) is directly linked to the initiation and progression of various autoimmune diseases. To determine the Treg-independent impact of NRF2 activation on autoimmune inflammation, we examined scurfy (Sf) mice, which are deficient in Tregs and succumb to severe multiorgan inflammation by 4 weeks of age. We found that systemic activation of NRF2 by Keap1 (Kelch-like ECH-associated protein 1) knockdown ameliorated tissue inflammation and lethality in Sf mice. Activated T cells and their cytokine production were accordingly decreased by Keap1 knockdown. In contrast, NRF2 activation through cell lineage-specific Keap1 disruption (i.e., in T cells, myeloid cells, and dendritic cells) achieved only partial or no improvement in the inflammatory status of Sf mice. Our results indicate that systemic activation of NRF2 suppresses effector T cell activities independently of Tregs and that NRF2 activation in multiple cell lineages appears to be required for sufficient anti-inflammatory effects. This study emphasizes the possible therapeutic application of NRF2 inducers in autoimmune diseases that are accompanied by Treg dysfunction.
Collapse
|
48
|
Dambuza IM, Levitz SM, Netea MG, Brown GD. Fungal Recognition and Host Defense Mechanisms. Microbiol Spectr 2017; 5:10.1128/microbiolspec.funk-0050-2016. [PMID: 28752813 PMCID: PMC11687525 DOI: 10.1128/microbiolspec.funk-0050-2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Indexed: 02/06/2023] Open
Abstract
Fungi have emerged as premier opportunistic microbes of the 21st century, having a considerable impact on human morbidity and mortality. The huge increase in incidence of these diseases is largely due to the HIV pandemic and use of immunosuppressive therapies, underscoring the importance of the immune system in defense against fungi. This article will address how the mammalian immune system recognizes and mounts a defense against medically relevant fungal species.
Collapse
Affiliation(s)
- I M Dambuza
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB252ZD, United Kingdom
| | - S M Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - M G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - G D Brown
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB252ZD, United Kingdom
| |
Collapse
|
49
|
Tung KSK, Harakal J, Qiao H, Rival C, Li JCH, Paul AGA, Wheeler K, Pramoonjago P, Grafer CM, Sun W, Sampson RD, Wong EWP, Reddi PP, Deshmukh US, Hardy DM, Tang H, Cheng CY, Goldberg E. Egress of sperm autoantigen from seminiferous tubules maintains systemic tolerance. J Clin Invest 2017; 127:1046-1060. [PMID: 28218625 DOI: 10.1172/jci89927] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/21/2016] [Indexed: 12/29/2022] Open
Abstract
Autoimmune responses to meiotic germ cell antigens (MGCA) that are expressed on sperm and testis occur in human infertility and after vasectomy. Many MGCA are also expressed as cancer/testis antigens (CTA) in human cancers, but the tolerance status of MGCA has not been investigated. MGCA are considered to be uniformly immunogenic and nontolerogenic, and the prevailing view posits that MGCA are sequestered behind the Sertoli cell barrier in seminiferous tubules. Here, we have shown that only some murine MGCA are sequestered. Nonsequestered MCGA (NS-MGCA) egressed from normal tubules, as evidenced by their ability to interact with systemically injected antibodies and form localized immune complexes outside the Sertoli cell barrier. NS-MGCA derived from cell fragments that were discarded by spermatids during spermiation. They egressed as cargo in residual bodies and maintained Treg-dependent physiological tolerance. In contrast, sequestered MGCA (S-MGCA) were undetectable in residual bodies and were nontolerogenic. Unlike postvasectomy autoantibodies, which have been shown to mainly target S-MGCA, autoantibodies produced by normal mice with transient Treg depletion that developed autoimmune orchitis exclusively targeted NS-MGCA. We conclude that spermiation, a physiological checkpoint in spermatogenesis, determines the egress and tolerogenicity of MGCA. Our findings will affect target antigen selection in testis and sperm autoimmunity and the immune responses to CTA in male cancer patients.
Collapse
|
50
|
Perri V, Gianchecchi E, Scarpa R, Valenzise M, Rosado MM, Giorda E, Crinò A, Cappa M, Barollo S, Garelli S, Betterle C, Fierabracci A. Altered B cell homeostasis and Toll-like receptor 9-driven response in patients affected by autoimmune polyglandular syndrome Type 1: Altered B cell phenotype and dysregulation of the B cell function in APECED patients. Immunobiology 2017; 222:372-383. [PMID: 27622939 DOI: 10.1016/j.imbio.2016.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/07/2016] [Indexed: 01/02/2023]
Abstract
APECED is a T-cell mediated disease with increased frequencies of CD8+ effector and reduction of FoxP3+ T regulatory cells. Antibodies against affected organs and neutralizing to cytokines are found in the peripheral blood. The contribution of B cells to multiorgan autoimmunity in Aire-/- mice was reported opening perspectives on the utility of anti-B cell therapy. We aimed to analyse the B cell phenotype of APECED patients compared to age-matched controls. FACS analysis was conducted on PBMC in basal conditions and following CpG stimulation. Total B and switched memory (SM) B cells were reduced while IgM memory were increased in patients. In those having more than 15 years from the first clinical manifestation the defect included also mature and transitional B cells; total memory B cells were increased, while SM were unaffected. In patients with shorter disease duration, total B cells were unaltered while SM and IgM memory behaved as in the total group. A defective B cell proliferation was detected after 4day-stimulation. In conclusion APECED patients show, in addition to a significant alteration of the B cell phenotype, a dysregulation of the B cell function involving peripheral innate immune mechanisms particularly those with longer disease duration.
Collapse
Affiliation(s)
- Valentina Perri
- Immuno-Infectivology and Clinical Trials Research Area, Children's Hospital Bambino Gesù, Rome, Italy
| | - Elena Gianchecchi
- Immuno-Infectivology and Clinical Trials Research Area, Children's Hospital Bambino Gesù, Rome, Italy
| | - Riccardo Scarpa
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Mariella Valenzise
- Unit of Pediatrics, Department of Pediatric Sciences, University of Messina, Messina, Italy
| | | | - Ezio Giorda
- Research Laboratories, Children's Hospital Bambino Gesù, Rome, Italy
| | - Antonino Crinò
- Endocrinology Department, Children's Hospital Bambino Gesù, Rome, Italy
| | - Marco Cappa
- Endocrinology Department, Children's Hospital Bambino Gesù, Rome, Italy
| | - Susi Barollo
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Silvia Garelli
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Corrado Betterle
- Endocrine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Alessandra Fierabracci
- Immuno-Infectivology and Clinical Trials Research Area, Children's Hospital Bambino Gesù, Rome, Italy.
| |
Collapse
|