1
|
Klein L, Petrozziello E. Antigen presentation for central tolerance induction. Nat Rev Immunol 2024:10.1038/s41577-024-01076-8. [PMID: 39294277 DOI: 10.1038/s41577-024-01076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/20/2024]
Abstract
The extent of central T cell tolerance is determined by the diversity of self-antigens that developing thymocytes 'see' on thymic antigen-presenting cells (APCs). Here, focusing on insights from the past decade, we review the functional adaptations of medullary thymic epithelial cells, thymic dendritic cells and thymic B cells for the purpose of tolerance induction. Their distinct cellular characteristics range from unconventional phenomena, such as promiscuous gene expression or mimicry of peripheral cell types, to strategic positioning in distinct microenvironments and divergent propensities to preferentially access endogenous or exogenous antigen pools. We also discuss how 'tonic' inflammatory signals in the thymic microenvironment may extend the intrathymically visible 'self' to include autoantigens that are otherwise associated with highly immunogenic peripheral environments.
Collapse
Affiliation(s)
- Ludger Klein
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.
| | - Elisabetta Petrozziello
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| |
Collapse
|
2
|
Brad GF, Nicoară DM, Scutca AC, Bugi MA, Asproniu R, Olariu LG, Jugănaru I, Cristun LI, Mărginean O. Exploring Chronic Hypocalcemia: Insights into Autoimmune Polyglandular Syndrome Type 1-A Case Study and Literature Review. J Clin Med 2024; 13:2368. [PMID: 38673639 PMCID: PMC11051075 DOI: 10.3390/jcm13082368] [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: 03/13/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Hypocalcemia is a common occurrence in pediatric patients, attributed to various causes and presenting with diverse clinical manifestations. A prompt evaluation is necessary to determine its underlying cause, whether it presents acutely or chronically, and to tailor treatment based on its severity. Among the potential causes of chronic hypocalcemia, primary hypoparathyroidism stands out. The case of a seven-year-old male patient with hypocalcemia reported in this article serves as an illustration, wherein targeted next-generation sequencing revealed a homozygous p.R257X mutation in the AIRE gene, indicative of autoimmune polyendocrine syndrome type 1 (APS-1). It poses challenges due to its multisystemic nature and involvement of specific autoantibodies, often leading to underdiagnosis, owing to its rarity, varied manifestations, and incomplete penetrance. A comprehensive review of the APS-1 literature was conducted to provide insights into the clinical manifestations, genetic spectrum, potential immunological mechanisms, and current medical strategies. Additionally, the recognition of AIRE gene mutations is crucial for facilitating genetic diagnosis, prognosis, and potential treatment strategies for APS-1. The management of such cases involves individualized approaches to treatment, regular monitoring, medication adjustments, and the early identification of associated conditions.
Collapse
Affiliation(s)
- Giorgiana-Flavia Brad
- Department XI Pediatrics, Discipline I Pediatrics, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania; (G.-F.B.); (A.-C.S.); (R.A.); (L.-G.O.); (I.J.); (O.M.)
- 1st Department of Pediatrics, Children’s Emergency Hospital ‘Louis Turcanu’, 300011 Timisoara, Romania;
| | - Delia-Maria Nicoară
- Department XI Pediatrics, Discipline I Pediatrics, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania; (G.-F.B.); (A.-C.S.); (R.A.); (L.-G.O.); (I.J.); (O.M.)
| | - Alexandra-Cristina Scutca
- Department XI Pediatrics, Discipline I Pediatrics, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania; (G.-F.B.); (A.-C.S.); (R.A.); (L.-G.O.); (I.J.); (O.M.)
- 1st Department of Pediatrics, Children’s Emergency Hospital ‘Louis Turcanu’, 300011 Timisoara, Romania;
| | - Meda-Ada Bugi
- 1st Department of Pediatrics, Children’s Emergency Hospital ‘Louis Turcanu’, 300011 Timisoara, Romania;
- Research Center for Disturbances of Growth and Development in Children BELIVE, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania
| | - Raluca Asproniu
- Department XI Pediatrics, Discipline I Pediatrics, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania; (G.-F.B.); (A.-C.S.); (R.A.); (L.-G.O.); (I.J.); (O.M.)
- 1st Department of Pediatrics, Children’s Emergency Hospital ‘Louis Turcanu’, 300011 Timisoara, Romania;
| | - Laura-Gratiela Olariu
- Department XI Pediatrics, Discipline I Pediatrics, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania; (G.-F.B.); (A.-C.S.); (R.A.); (L.-G.O.); (I.J.); (O.M.)
- 1st Department of Pediatrics, Children’s Emergency Hospital ‘Louis Turcanu’, 300011 Timisoara, Romania;
| | - Iulius Jugănaru
- Department XI Pediatrics, Discipline I Pediatrics, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania; (G.-F.B.); (A.-C.S.); (R.A.); (L.-G.O.); (I.J.); (O.M.)
- 1st Department of Pediatrics, Children’s Emergency Hospital ‘Louis Turcanu’, 300011 Timisoara, Romania;
- Research Center for Disturbances of Growth and Development in Children BELIVE, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania
| | - Lucian-Ioan Cristun
- Ph.D. School Department, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania;
| | - Otilia Mărginean
- Department XI Pediatrics, Discipline I Pediatrics, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania; (G.-F.B.); (A.-C.S.); (R.A.); (L.-G.O.); (I.J.); (O.M.)
- 1st Department of Pediatrics, Children’s Emergency Hospital ‘Louis Turcanu’, 300011 Timisoara, Romania;
- Research Center for Disturbances of Growth and Development in Children BELIVE, ‘Victor Babes’ University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania
| |
Collapse
|
3
|
Källberg E, Mehmeti-Ajradini M, Björk Gunnarsdottir F, Göransson M, Bergenfelz C, Allaoui Fredriksson R, Hagerling C, Johansson ME, Welinder C, Jirström K, Leandersson K. AIRE is expressed in breast cancer TANs and TAMs to regulate the extrinsic apoptotic pathway and inflammation. J Leukoc Biol 2024; 115:664-678. [PMID: 38060995 DOI: 10.1093/jleuko/qiad152] [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: 08/15/2023] [Revised: 11/02/2023] [Accepted: 11/19/2023] [Indexed: 04/02/2024] Open
Abstract
The autoimmune regulator (AIRE) is a transcriptional regulator expressed in the thymus and is necessary for maintaining immunological self-tolerance. Extrathymic AIRE expression is rare, and a role for AIRE in tumor-associated innate immune cells has not yet been established. In this study, we show that AIRE is expressed in human pro-tumor neutrophils. In breast cancer, AIRE was primarily located to tumor-associated neutrophils (TANs), and to a lesser extent to tumor-associated macrophages (TAMs) and tumor cells. Expression of AIRE in TAN/TAMs, but not in cancer cells, was associated with an adverse prognosis. We show that the functional role for AIRE in neutrophils and macrophages is to regulate expression of immune mediators and the extrinsic apoptotic pathway involving the Fas/TNFR death receptors and cathepsin G. Here, we propose that the role for AIRE in TAN/TAMs in breast tumors is to regulate cell death and inflammation, thus promoting tumor progression.
Collapse
Affiliation(s)
- Eva Källberg
- Cancer Immunology, Department of Translational Medicine, Lund University, Jan Waldenströmsg 35, 214 28 Malmö, Sweden
| | - Meliha Mehmeti-Ajradini
- Cancer Immunology, Department of Translational Medicine, Lund University, Jan Waldenströmsg 35, 214 28 Malmö, Sweden
| | - Frida Björk Gunnarsdottir
- Cancer Immunology, Department of Translational Medicine, Lund University, Jan Waldenströmsg 35, 214 28 Malmö, Sweden
| | - Marcus Göransson
- Cancer Immunology, Department of Translational Medicine, Lund University, Jan Waldenströmsg 35, 214 28 Malmö, Sweden
| | - Caroline Bergenfelz
- Cancer Immunology, Department of Translational Medicine, Lund University, Jan Waldenströmsg 35, 214 28 Malmö, Sweden
| | - Roni Allaoui Fredriksson
- Cancer Immunology, Department of Translational Medicine, Lund University, Jan Waldenströmsg 35, 214 28 Malmö, Sweden
| | - Catharina Hagerling
- Cancer Immunology, Department of Translational Medicine, Lund University, Jan Waldenströmsg 35, 214 28 Malmö, Sweden
| | - Martin E Johansson
- Sahlgrenska Center for Cancer Research, Department of Biomedicine, Vasaparken Universitetsplatsen 1, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Charlotte Welinder
- Mass Spectrometry, Department for Clinical Sciences, Lund University, Sölvegatan 19, 221 84 Lund, Sweden
| | - Karin Jirström
- Oncology and Therapeutic Pathology, Department of Clinical Sciences Lund, Lund University, Sölvegatan 19, 221 84 Lund, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department of Translational Medicine, Lund University, Jan Waldenströmsg 35, 214 28 Malmö, Sweden
| |
Collapse
|
4
|
Laan M, Salumets A, Klein A, Reintamm K, Bichele R, Peterson H, Peterson P. Post-Aire Medullary Thymic Epithelial Cells and Hassall's Corpuscles as Inducers of Tonic Pro-Inflammatory Microenvironment. Front Immunol 2021; 12:635569. [PMID: 33868260 PMCID: PMC8050345 DOI: 10.3389/fimmu.2021.635569] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/05/2021] [Indexed: 11/13/2022] Open
Abstract
While there is convincing evidence on the role of Aire-positive medullary thymic epithelial cells (mTEC) in the induction of central tolerance, the nature and function of post-Aire mTECs and Hassall's corpuscles have remained enigmatic. Here we summarize the existing data on these late stages of mTEC differentiation with special focus on their potential to contribute to central tolerance induction by triggering the unique pro-inflammatory microenvironment in the thymus. In order to complement the existing evidence that has been obtained from mouse models, we performed proteomic analysis on microdissected samples from human thymic medullary areas at different differentiation stages. The analysis confirms that at the post-Aire stages, the mTECs lose their nuclei but maintain machinery required for translation and exocytosis and also upregulate proteins specific to keratinocyte differentiation and cornification. In addition, at the late stages of differentiation, the human mTECs display a distinct pro-inflammatory signature, including upregulation of the potent endogenous TLR4 agonist S100A8/S100A9. Collectively, the study suggests a novel mechanism by which the post-Aire mTECs and Hassall's corpuscles contribute to the thymic microenvironment with potential cues on the induction of central tolerance.
Collapse
Affiliation(s)
- Martti Laan
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Ahto Salumets
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Institute of Computer Science, Faculty of Science and Technology, University of Tartu, Tartu, Estonia
| | - Annabel Klein
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kerli Reintamm
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Rudolf Bichele
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Hedi Peterson
- Institute of Computer Science, Faculty of Science and Technology, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
5
|
Evaluation of silent information regulator T (SIRT) 1 and Forkhead Box O (FOXO) transcription factor 1 and 3a genes in glaucoma. Mol Biol Rep 2020; 47:9337-9344. [PMID: 33200312 DOI: 10.1007/s11033-020-05994-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 11/06/2020] [Indexed: 12/16/2022]
Abstract
Analysis of the reactive oxygen species (ROS)-detoxifying biomarkers may elucidate the mitochondrial dysfunction in glaucoma pathogenesis. Therefore, we purposed to investigate the effects of ROS-detoxifying molecules including Silent Information Regulator T1 (SIRT1) and Forkhead Box O 1 (FOXO1) and 3a (FOXO3a) transcription factors in patients with glaucoma. Our analyses included 20 eyes from patients with primary open-angle glaucoma (POAG) and 20 eyes from patients with pseudoexfoliation glaucoma (PXG) who were scheduled for trabeculectomy. After extraction of total RNA from trabecular meshwork tissue, we compared the levels of SIRT1, FOXO1and FOXO3a genes in the oxidative pathway with the level of glyceraldehyde-3 phosphate dehydrogenase (GAPDH), the reference gene, using real-time polymerase chain reaction. Relative gene expression was calculated using the threshold cycle (2-ΔΔCT) method. We observed similarly reduced expression levels of SIRT1, FOXO1, and FOXO3a genes versus GAPDH among patient groups (p = 0.40; p = 0.56; p = 0.35, respectively). This is the first study to identify the role of SIRT1 and FOXOs in human TM with glaucoma. Relative expression levels of SIRT1, FOXO1, and FOXO3a genes versus a control gene (GAPDH) were decreased in POAG and PXG groups. Our results show that SIRT1and FOXOs (1-3a) deserve special attention in the pathogenesis of glaucoma.
Collapse
|
6
|
Zhang L, Zhang H, Yang S. Cytosolic TaGAPC2 Enhances Tolerance to Drought Stress in Transgenic Arabidopsis Plants. Int J Mol Sci 2020; 21:ijms21207499. [PMID: 33053684 PMCID: PMC7590034 DOI: 10.3390/ijms21207499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 11/16/2022] Open
Abstract
Drought is a major natural disaster that seriously affects agricultural production, especially for winter wheat in boreal China. As functional proteins, the functions and mechanisms of glyceraldehyde-3-phosphate dehydrogenase in cytoplasm (GAPCs) have remained little investigated in wheat subjected to adverse environmental conditions. In this study, we cloned and characterized a GAPC isoform TaGAPC2 in wheat. Over-expression of TaGApC2-6D in Arabidopsis led to enhanced root length, reduced reactive oxygen species (ROS) production, and elevated drought tolerance. In addition, the dual-luciferase assays showed that TaWRKY28/33/40/47 could positively regulate the expression of TaGApC2-6A and TaGApC2-6D. Further results of the yeast two-hybrid system and bimolecular fluorescence complementation assay (BiFC) demonstrate that TaPLDδ, an enzyme producing phosphatidic acid (PA), could interact with TaGAPC2-6D in plants. These results demonstrate that TaGAPC2 regulated by TaWRKY28/33/40/47 plays a crucial role in drought tolerance, which may influence the drought stress conditions via interaction with TaPLDδ. In conclusion, our results establish a new positive regulation mechanism of TaGAPC2 that helps wheat fine-tune its drought response.
Collapse
|
7
|
Gong B, Wang X, Li B, Li Y, Lu R, Zhang K, Li B, Ma Y, Li Y. miR-205-5p inhibits thymic epithelial cell proliferation via FA2H-TFAP2A feedback regulation in age-associated thymus involution. Mol Immunol 2020; 122:173-185. [PMID: 32371259 DOI: 10.1016/j.molimm.2020.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/02/2020] [Accepted: 04/13/2020] [Indexed: 12/28/2022]
Abstract
Thymic epithelial cells (TECs) are essential regulators of T cell development and selection. microRNAs (miRNAs) play critical roles in regulating TECs proliferation during thymus involution. miR-205-5p is highly expressed in TECs and increases with age. However, the function and potential mechanism of miR-205-5p in TECs are not clear. miRNA expression was profiled using TECs from male and female mice at 1 and 3 months old. A total of 325 differentially expressed miRNAs (DEMs) were detected at different ages in two sexes. 24 of the DEMs had the same trend between males and females. Among them, miR-205-5p had the highest fold change. Our results showed that the expression of miR-205-5p was dramatically increased in TECs from 1 to 9 months old mice. miR-205-5p mimic inhibited TECs proliferation. Moreover, we confirmed that Fa2h was the direct target gene of miR-205-5p and FA2H was significantly decreased in TECs with increased expression of miR-205-5p. Silencing of Fa2h inhibited TECs proliferation. Furthermore, we found that the expression of Tfap2a could be promoted by FA2H and that TFAP2A could interact with miR-205-5p in TECs. Overall, miR-205-5p is an important regulator of TECs proliferation and regulates age-associated thymus involution via the miR-205-5p-FA2H-TFAP2A feedback regulatory circuit. miR-205-5p might act as a potential biomarker in TECs for age-related thymus involution.
Collapse
Affiliation(s)
- Bishuang Gong
- College of Veterinary Medicine, South China Agricultural University, China
| | - Xintong Wang
- College of Veterinary Medicine, South China Agricultural University, China
| | - Boning Li
- the Department of Cardiology, Shenzhen Children's Hospital, Shenzhen, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, China
| | - Rui Lu
- College of Veterinary Medicine, South China Agricultural University, China
| | - Kaizhao Zhang
- College of Veterinary Medicine, South China Agricultural University, China
| | - Bingxin Li
- College of Veterinary Medicine, South China Agricultural University, China
| | - Yongjiang Ma
- College of Veterinary Medicine, South China Agricultural University, China.
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, China.
| |
Collapse
|
8
|
Melo‐Lima BL, Poras I, Passos GA, Carosella ED, Donadi EA, Moreau P. The Autoimmune Regulator (Aire) transactivates HLA-G gene expression in thymic epithelial cells. Immunology 2019; 158:121-135. [PMID: 31322727 PMCID: PMC6742766 DOI: 10.1111/imm.13099] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 07/11/2019] [Indexed: 12/12/2022] Open
Abstract
The Autoimmune Regulator (Aire) protein coordinates the negative selection of developing thymocytes by inducing the expression of hundreds of tissue-specific antigens within the thymic medulla, which is also a primary site of the expression of the immune checkpoint HLA-G molecule. Considering the immunomodulatory properties of Aire and HLA-G, and considering that the role of the constitutive thymus expression of HLA-G has not been elucidated, we studied the effect of AIRE cDNA transfection on HLA-G expression in 4D6 thymic cells and in the HLA-G-positive JEG-3 choriocarcinoma cells. Aire promoted the transactivation of HLA-G gene by increasing the overall transcription, inducing the transcription of at least G1 and G2/G4 isoforms, and incrementing the occurrence and distribution of intracellular HLA-G protein solely in 4D6 thymic cells. Luciferase-based assays and chromatin immunoprecipitation experiments performed in 4D6 cells revealed that Aire targeted at least two regions within the 5'-untranslated regulatory region (5'-URR) extending 1·4 kb from the first ATG initiation codon. The interaction occurs independently of three putative Aire-binding sites. These results indicate that the Aire-induced upregulation of HLA-G in thymic cells is likely to act through the interaction of Aire with specific HLA-G 5'-URR DNA-binding factors. Such a multimeric transcriptional complex might operate in the thymus during the process of promiscuous gene expression.
Collapse
Affiliation(s)
- Breno Luiz Melo‐Lima
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
- Division of Clinical ImmunologyDepartment of MedicineRibeirao Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Isabelle Poras
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| | - Geraldo Aleixo Passos
- Molecular Immunogenetics GroupDepartment of GeneticsRibeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Edgardo D. Carosella
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| | - Eduardo Antonio Donadi
- Division of Clinical ImmunologyDepartment of MedicineRibeirao Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Philippe Moreau
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| |
Collapse
|
9
|
Michel C, Miller CN, Küchler R, Brors B, Anderson MS, Kyewski B, Pinto S. Revisiting the Road Map of Medullary Thymic Epithelial Cell Differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:3488-3503. [PMID: 28993517 DOI: 10.4049/jimmunol.1700203] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 09/08/2017] [Indexed: 11/19/2022]
Abstract
The basic two-step terminal differentiation model of the medullary thymic epithelial cell (mTEC) lineage from immature MHC class II (MHCII)lo to mature MHCIIhi mTECs has recently been extended to include a third stage, namely the post-Aire MHCIIlo subset as identified by lineage-tracing models. However, a suitable surface marker distinguishing the phenotypically overlapping pre- from the post-Aire MHCIIlo stage has been lacking. In this study, we introduce the lectin Tetragonolobus purpureas agglutinin (TPA) as a novel cell surface marker that allows for such delineation. Based on our data, we derived the following sequence of mTEC differentiation: TPAloMHCIIlo → TPAloMHCIIhi → TPAhiMHCIIhi → TPAhiMHCIIlo Surprisingly, in the steady-state postnatal thymus TPAloMHCIIlo pre-Aire rather than terminally differentiated post-Aire TPAhiMHCIIlo mTECs were marked for apoptosis at an exceptionally high rate of ∼70%. Hence, only the minor cycling fraction of the MHCIIlo subset (<20%) potentially qualified as mTEC precursors. FoxN1 expression inversely correlated with the fraction of slow cycling and apoptotic cells within the four TPA subsets. TPA also further subdivided human mTECs, although with different subset distribution. Our revised road map emphazises close parallels of terminal mTEC development with that of skin, undergoing an alternative route of cell death, namely cornification rather than apoptosis. The high rate of apoptosis in pre-Aire MHCIIlo mTECs points to a "quality control" step during early mTEC differentiation.
Collapse
Affiliation(s)
- Chloé Michel
- Division of Developmental Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Corey N Miller
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143
| | - Rita Küchler
- Division of Developmental Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center, Heidelberg 69120, Germany
- National Center for Tumor Diseases, Heidelberg 69120, Germany; and
- German Cancer Consortium, Heidelberg 69120, Germany
| | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143
| | - Bruno Kyewski
- Division of Developmental Immunology, German Cancer Research Center, Heidelberg 69120, Germany;
| | - Sheena Pinto
- Division of Developmental Immunology, German Cancer Research Center, Heidelberg 69120, Germany;
| |
Collapse
|
10
|
A critical epithelial survival axis regulated by MCL-1 maintains thymic function in mice. Blood 2017; 130:2504-2515. [PMID: 28972012 DOI: 10.1182/blood-2017-03-771576] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 09/21/2017] [Indexed: 12/15/2022] Open
Abstract
T-cell differentiation is governed by interactions with thymic epithelial cells (TECs) and defects in this process undermine immune function and tolerance. To uncover new strategies to restore thymic function and adaptive immunity in immunodeficiency, we sought to determine the molecular mechanisms that control life and death decisions in TECs. Guided by gene expression profiling, we created mouse models that specifically deleted prosurvival genes in TECs. We found that although BCL-2 and BCL-XL were dispensable for TEC homeostasis, MCL-1 deficiency impacted on TECs as early as embryonic day 15.5, resulting in early thymic atrophy and T-cell lymphopenia, with near complete loss of thymic tissue by 2 months of age. MCL-1 was not necessary for TEC differentiation but was continually required for the survival of mature cortical and medullary TECs and the maintenance of thymic architecture. A screen of TEC trophic factors in organ cultures showed that epidermal growth factor upregulated MCL-1 via MAPK/ERK kinase activity, providing a molecular mechanism for the support of TEC survival. This signaling axis governing TEC survival and thymic function represents a new target for strategies for thymic protection and regeneration.
Collapse
|
11
|
Guha M, Saare M, Maslovskaja J, Kisand K, Liiv I, Haljasorg U, Tasa T, Metspalu A, Milani L, Peterson P. DNA breaks and chromatin structural changes enhance the transcription of autoimmune regulator target genes. J Biol Chem 2017; 292:6542-6554. [PMID: 28242760 PMCID: PMC5399106 DOI: 10.1074/jbc.m116.764704] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/30/2017] [Indexed: 12/22/2022] Open
Abstract
The autoimmune regulator (AIRE) protein is the key factor in thymic negative selection of autoreactive T cells by promoting the ectopic expression of tissue-specific genes in the thymic medullary epithelium. Mutations in AIRE cause a monogenic autoimmune disease called autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. AIRE has been shown to promote DNA breaks via its interaction with topoisomerase 2 (TOP2). In this study, we investigated topoisomerase-induced DNA breaks and chromatin structural alterations in conjunction with AIRE-dependent gene expression. Using RNA sequencing, we found that inhibition of TOP2 religation activity by etoposide in AIRE-expressing cells had a synergistic effect on genes with low expression levels. AIRE-mediated transcription was not only enhanced by TOP2 inhibition but also by the TOP1 inhibitor camptothecin. The transcriptional activation was associated with structural rearrangements in chromatin, notably the accumulation of γH2AX and the exchange of histone H1 with HMGB1 at AIRE target gene promoters. In addition, we found the transcriptional up-regulation to co-occur with the chromatin structural changes within the genomic cluster of carcinoembryonic antigen-like cellular adhesion molecule genes. Overall, our results suggest that the presence of AIRE can trigger molecular events leading to an altered chromatin landscape and the enhanced transcription of low-expressed genes.
Collapse
Affiliation(s)
- Mithu Guha
- From the Molecular Pathology, Institute of Biomedical and Translational Medicine
| | - Mario Saare
- From the Molecular Pathology, Institute of Biomedical and Translational Medicine
| | - Julia Maslovskaja
- From the Molecular Pathology, Institute of Biomedical and Translational Medicine
| | - Kai Kisand
- From the Molecular Pathology, Institute of Biomedical and Translational Medicine
| | - Ingrid Liiv
- From the Molecular Pathology, Institute of Biomedical and Translational Medicine
| | - Uku Haljasorg
- From the Molecular Pathology, Institute of Biomedical and Translational Medicine
| | | | - Andres Metspalu
- Estonian Genome Center, and
- Institute of Molecular and Cell Biology, University of Tartu, Tartu 50411, Estonia
| | | | - Pärt Peterson
- From the Molecular Pathology, Institute of Biomedical and Translational Medicine,
| |
Collapse
|
12
|
Comparative proteomic analysis of liver antioxidant mechanisms in Megalobrama amblycephala stimulated with dietary emodin. Sci Rep 2017; 7:40356. [PMID: 28084435 PMCID: PMC5233964 DOI: 10.1038/srep40356] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 12/05/2016] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress is a toxicological endpoint that correlates with the nutrition status of fish through cellular damage, inflammation, and apoptosis. In order to understand the antioxidant mechanism induced by dietary emodin in Megalobrama amblycephala liver, a comparative proteomic analysis was performed to investigate the proteome alteration under emodin administration. 27 altered protein spots were separated under 30 mg kg-1 emodin stimulation based on 2-DE, and were all successfully identified using MALDI-TOF/TOF, representing 17 unique proteins. These proteins were functionally classified into antioxidant, metabolism, cytoskeleton, chaperone, signal transduction and cofactor groups. Network interaction and Gene Ontology annotation indicated 10 unique proteins were closely related to antioxidation and directly regulated by each other. Compared with the control group, administration of 30 mg kg-1 emodin significantly increased the antioxidant-related mRNA expressions of GPx1, GSTm and HSP70, but decreased the mRNA expressions of GAPDH and Sord, which was consistent with the protein expression. Nevertheless, Pgk1 and Aldh8a1 were up- and down-regulated, and ALDOB was down- and up-regulated at the mRNA and protein levels, respectively. These results revealed that the altered proteins enhanced antioxidation via complex regulatory mechanisms, and 30 mg kg-1 emodin was a suitable immunostimulant for M. amblycephala.
Collapse
|
13
|
Bianchi F, Sommariva M, De Cecco L, Triulzi T, Romero-Cordoba S, Tagliabue E, Sfondrini L, Balsari A. Expression and prognostic significance of the autoimmune regulator gene in breast cancer cells. Cell Cycle 2016; 15:3220-3229. [PMID: 27753538 PMCID: PMC5176139 DOI: 10.1080/15384101.2016.1241918] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/09/2016] [Accepted: 09/21/2016] [Indexed: 10/20/2022] Open
Abstract
The autoimmune regulator gene (AIRE) plays a fundamental role in tolerance by promoting the expression of tissue-specific antigens in medullary thymic epithelial cells (mTECs). Recently, AIRE expression was detected also in human keratinocytes and in tumors originating in stratified epithelia. Here, we tested whether AIRE is expressed in cancer cells. We analyzed AIRE expression in cancer cases from The Cancer Genome Atlas (TCGA) RNA-seq dataset and we found association with better outcome. AIRE protein expression was verified by immunohistochemistry in a cohort of 39 human breast cancer specimens and its prognostic relevance was confirmed in microarray-based gene expression data set NKI-295 and KM-Plotter. Both in the RNA-seq and gene expression datasets analyzed, AIRE expression was an independent strong prognostic factor for relapse-free survival (RFS), particularly in estrogen receptor-positive tumors. Enrichment of translation-related pathways was observed in AIRE-expressing tumors by Ingenuity Pathway Analysis and a significant increase of cells in G1 phase and activation of caspase cascades was induced by AIRE transfection in breast cancer luminal cell lines, suggesting that AIRE-induced over-translation of proteins lead to cycle arrest and apoptosis. These data are the first to identify AIRE expression in breast cancer and an association with prognosis.
Collapse
Affiliation(s)
- Francesca Bianchi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Loris De Cecco
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics Core Facility, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sandra Romero-Cordoba
- Oncogenomics Laboratory, National Institute of Genomics Medicine, Mexico City, Mexico
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Andrea Balsari
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
14
|
Radhakrishnan K, Bhagya KP, Kumar AT, Devi AN, Sengottaiyan J, Kumar PG. Autoimmune Regulator (AIRE) Is Expressed in Spermatogenic Cells, and It Altered the Expression of Several Nucleic-Acid-Binding and Cytoskeletal Proteins in Germ Cell 1 Spermatogonial (GC1-spg) Cells. Mol Cell Proteomics 2016; 15:2686-98. [PMID: 27281783 PMCID: PMC4974344 DOI: 10.1074/mcp.m115.052951] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 05/24/2016] [Indexed: 11/06/2022] Open
Abstract
Autoimmune regulator (AIRE) is a gene associated with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED). AIRE is expressed heavily in the thymic epithelial cells and is involved in maintaining self-tolerance through regulating the expression of tissue-specific antigens. The testes are the most predominant extrathymic location where a heavy expression of AIRE is reported. Homozygous Aire-deficient male mice were infertile, possibly due to impaired spermatogenesis, deregulated germ cell apoptosis, or autoimmunity. We report that AIRE is expressed in the testes of neonatal, adolescent, and adult mice. AIRE expression was detected in glial cell derived neurotrophic factor receptor alpha (GFRα)(+) (spermatogonia), GFRα(-)/synaptonemal complex protein (SCP3)(+) (meiotic), and GFRα(-)/Phosphoglycerate kinase 2 (PGK2)(+) (postmeiotic) germ cells in mouse testes. GC1-spg, a germ-cell-derived cell line, did not express AIRE. Retinoic acid induced AIRE expression in GC1-spg cells. Ectopic expression of AIRE in GC1-spg cells using label-free LC-MS/MS identified a total of 371 proteins that were differentially expressed. 100 proteins were up-regulated, and 271 proteins were down-regulated. Data are available via ProteomeXchange with identifier PXD002511. Functional analysis of the differentially expressed proteins showed increased levels of various nucleic-acid-binding proteins and transcription factors and a decreased level of various cytoskeletal and structural proteins in the AIRE overexpressing cells as compared with the empty vector-transfected controls. The transcripts of a select set of the up-regulated proteins were also elevated. However, there was no corresponding decrease in the mRNA levels of the down-regulated set of proteins. Molecular function network analysis indicated that AIRE influenced gene expression in GC1-spg cells by acting at multiple levels, including transcription, translation, RNA processing, protein transport, protein localization, and protein degradation, thus setting the foundation in understanding the functional role of AIRE in germ cell biology.
Collapse
Affiliation(s)
- Karthika Radhakrishnan
- From the §Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Kongattu P Bhagya
- From the §Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Anil Tr Kumar
- From the §Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Anandavalli N Devi
- From the §Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Jeeva Sengottaiyan
- From the §Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Pradeep G Kumar
- From the §Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| |
Collapse
|
15
|
Oh J, Shin JS. The Role of Dendritic Cells in Central Tolerance. Immune Netw 2015; 15:111-20. [PMID: 26140042 PMCID: PMC4486773 DOI: 10.4110/in.2015.15.3.111] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/26/2015] [Accepted: 06/02/2015] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) play a significant role in establishing self-tolerance through their ability to present self-antigens to developing T cells in the thymus. DCs are predominantly localized in the medullary region of thymus and present a broad range of self-antigens, which include tissue-restricted antigens expressed and transferred from medullary thymic epithelial cells, circulating antigens directly captured by thymic DCs through coticomedullary junction blood vessels, and peripheral tissue antigens captured and transported by peripheral tissue DCs homing to the thymus. When antigen-presenting DCs make a high affinity interaction with antigen-specific thymocytes, this interaction drives the interacting thymocytes to death, a process often referred to as negative selection, which fundamentally blocks the self-reactive thymocytes from differentiating into mature T cells. Alternatively, the interacting thymocytes differentiate into the regulatory T (Treg) cells, a distinct T cell subset with potent immune suppressive activities. The specific mechanisms by which thymic DCs differentiate Treg cells have been proposed by several laboratories. Here, we review the literatures that elucidate the contribution of thymic DCs to negative selection and Treg cell differentiation, and discusses its potential mechanisms and future directions.
Collapse
Affiliation(s)
- Jaehak Oh
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jeoung-Sook Shin
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
16
|
Smith RW, Wang J, Mothersill CE, Lee LEJ, Seymour CB. Proteomic responses in the gills of fathead minnows (Pimephales promelas, Rafinesque, 1820) after 6 months and 2 years of continuous exposure to environmentally relevant dietary226Ra. Int J Radiat Biol 2015; 91:248-56. [DOI: 10.3109/09553002.2014.988894] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
17
|
Zhai D, Chin K, Wang M, Liu F. Disruption of the nuclear p53-GAPDH complex protects against ischemia-induced neuronal damage. Mol Brain 2014; 7:20. [PMID: 24670206 PMCID: PMC3986870 DOI: 10.1186/1756-6606-7-20] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 02/28/2014] [Indexed: 12/24/2022] Open
Abstract
Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is conventionally considered a critical enzyme that involves in glycolysis for energy production. Recent previous studies have suggested that GAPDH is important in glutamate-induced neuronal excitotoxicity, while accumulated evidence also demonstrated that GAPDH nuclear translocation plays a critical role in cell death. However, the molecular mechanisms underlying this process remain largely unknown. In this study, we showed that GAPDH translocates to the nucleus in a Siah1-dependent manner upon glutamate stimulation. The nuclear GAPDH forms a protein complex with p53 and enhances p53 expression and phosphorylation. Disruption of the GAPDH-p53 interaction with an interfering peptide blocks glutamate-induced cell death and GAPDH-mediated up-regulation of p53 expression and phosphorylation. Furthermore, administration of the interfering peptide in vivo protects against ischemia-induced cell death in rats subjected to tMCAo. Our data suggest that the nuclear p53-GAPDH complex is important in regulating glutamate-mediated neuronal death and could serve as a potential therapeutic target for ischemic stroke treatment.
Collapse
Affiliation(s)
| | | | | | - Fang Liu
- Department of Neuroscience, Centre for Addiction and Mental Health, Clarke Division, 250 College Street, Toronto, Ontario M5T 1R8, Canada.
| |
Collapse
|
18
|
Thymic epithelial cell development and its dysfunction in human diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:206929. [PMID: 24672784 PMCID: PMC3929497 DOI: 10.1155/2014/206929] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 11/28/2013] [Indexed: 12/01/2022]
Abstract
Thymic epithelial cells (TECs) are the key components in thymic microenvironment for T cells development. TECs, composed of cortical and medullary TECs, are derived from a common bipotent progenitor and undergo a stepwise development controlled by multiple levels of signals to be functionally mature for supporting thymocyte development. Tumor necrosis factor receptor (TNFR) family members including the receptor activator for NFκB (RANK), CD40, and lymphotoxin β receptor (LTβR) cooperatively control the thymic medullary microenvironment and self-tolerance establishment. In addition, fibroblast growth factors (FGFs), Wnt, and Notch signals are essential for establishment of functional thymic microenvironment. Transcription factors Foxn1 and autoimmune regulator (Aire) are powerful modulators of TEC development, differentiation, and self-tolerance. Dysfunction in thymic microenvironment including defects of TEC and thymocyte development would cause physiological disorders such as tumor, infectious diseases, and autoimmune diseases. In the present review, we will summarize our current understanding on TEC development and the underlying molecular signals pathways and the involvement of thymus dysfunction in human diseases.
Collapse
|
19
|
Perniola R, Musco G. The biophysical and biochemical properties of the autoimmune regulator (AIRE) protein. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:326-37. [PMID: 24275490 DOI: 10.1016/j.bbadis.2013.11.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 11/11/2013] [Accepted: 11/18/2013] [Indexed: 01/20/2023]
Abstract
AIRE (for autoimmune regulator) is a multidomain protein that performs a fundamental function in the thymus and possibly in the secondary lymphoid organs: the regulation, especially in the sense of activation, of the process of gene transcription in cell lines deputed to the presentation of self-antigens to the maturing T lymphocytes. The apoptosis of the elements bearing T-cell receptors with critical affinity for the exhibited self-antigens prevents the escape of autoreactive clones and represents a simple and efficient mechanism of deletional self-tolerance. However, AIRE action relies on an articulated complex of biophysical and biochemical properties, in most cases attributable to single subspecialized domains. Here a thorough review of the matter is presented, with a privileged look at the pathogenic changes of AIRE that interfere with such properties and lead to the impairment in its chief function.
Collapse
Affiliation(s)
- Roberto Perniola
- Department of Pediatrics - Neonatal Intensive Care, V. Fazzi Regional Hospital, Piazza F. Muratore, I-73100, Lecce, Italy.
| | - Giovanna Musco
- Biomolecular NMR Laboratory, Center of Translational Genomics and Bioinformatics, Dulbecco Telethon Institute at San Raffaele Scientific Institute, Via Olgettina 58, I-20132, Milan, Italy.
| |
Collapse
|
20
|
Collado JA, Guitart C, Ciudad MT, Alvarez I, Jaraquemada D. The Repertoires of Peptides Presented by MHC-II in the Thymus and in Peripheral Tissue: A Clue for Autoimmunity? Front Immunol 2013; 4:442. [PMID: 24381570 PMCID: PMC3865459 DOI: 10.3389/fimmu.2013.00442] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 11/26/2013] [Indexed: 11/13/2022] Open
Abstract
T-cell tolerance to self-antigens is established in the thymus through the recognition by developing thymocytes of self-peptide-MHC complexes and induced and maintained in the periphery. Efficient negative selection of auto-reactive T cells in the thymus is dependent on the in situ expression of both ubiquitous and tissue-restricted self-antigens and on the presentation of derived peptides. Weak or inadequate intrathymic expression of self-antigens increases the risk to generate an autoimmune-prone T-cell repertoire. Indeed, even small changes of self-antigen expression in the thymus affect negative selection and increase the predisposition to autoimmunity. Together with other mechanisms, tolerance is maintained in the peripheral lymphoid organs via the recognition by mature T cells of a similar set of self-peptides in homeostatic conditions. However, non-lymphoid peripheral tissue, where organ-specific autoimmunity takes place, often have differential functional processes that may lead to the generation of epitopes that are absent or non-presented in the thymus. These putative differences between peptides presented by MHC molecules in the thymus and in peripheral tissues might be a major key to the initiation and maintenance of autoimmune conditions.
Collapse
Affiliation(s)
- Javier A Collado
- Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Carolina Guitart
- Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - M Teresa Ciudad
- Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Iñaki Alvarez
- Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Dolores Jaraquemada
- Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona , Spain
| |
Collapse
|
21
|
Laan M, Peterson P. The many faces of aire in central tolerance. Front Immunol 2013; 4:326. [PMID: 24130560 PMCID: PMC3795325 DOI: 10.3389/fimmu.2013.00326] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/24/2013] [Indexed: 11/13/2022] Open
Abstract
Although the role that Autoimmune Regulator (Aire) plays in the induction of central tolerance is well known, the precise cellular and molecular mechanisms are still unclear and debated. In the prevailing view, Aire serves mainly as a direct inducer of tissue-specific antigens. However, there is a growing amount of evidence suggesting that Aire modulates the differentiation program of medullary thymic epithelial cells, which may directly contribute to the negative selection of self-reactive thymocytes. In addition, Aire has been shown to regulate the expression of many intrathymic chemokines that are required for the proper localization of thymocytes and dendritic cells, and thus are potentially important for direct and indirect self-antigen presentation in the thymic medulla. Further, recent evidence suggests that the induction of certain antigen-specific regulatory T-cells that translocate to tumors and peripheral tissues can be Aire dependent and may contribute to tissue-specific tolerance. This review summarizes the current understanding of the effects of Aire on these alternative mechanisms for the induction of Aire-induced central tolerance.
Collapse
Affiliation(s)
- Martti Laan
- Molecular Pathology, Faculty of Medicine, Institute of Biomedicine and Translational Medicine, Tartu University , Tartu , Estonia
| | | |
Collapse
|
22
|
Sun L, Luo H, Li H, Zhao Y. Thymic epithelial cell development and differentiation: cellular and molecular regulation. Protein Cell 2013; 4:342-55. [PMID: 23589020 DOI: 10.1007/s13238-013-3014-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/11/2013] [Indexed: 11/26/2022] Open
Abstract
Thymic epithelial cells (TECs) are one of the most important components in thymic microenvironment supporting thymocyte development and maturation. TECs, composed of cortical and medullary TECs, are derived from a common bipotent progenitor, mediating thymocyte positive and negative selections. Multiple levels of signals including intracellular signaling networks and cell-cell interaction are required for TEC development and differentiation. Transcription factors Foxn1 and autoimmune regulator (Aire) are powerful regulators promoting TEC development and differentiation. Crosstalks with thymocytes and other stromal cells for extrinsic signals like RANKL, CD40L, lymphotoxin, fibroblast growth factor (FGF) and Wnt are also definitely required to establish a functional thymic microenvironment. In this review, we will summarize our current understanding about TEC development and differentiation, and its underlying multiple signal pathways.
Collapse
Affiliation(s)
- Lina Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | |
Collapse
|
23
|
Desanti GE, Cowan J, Baik S, Parnell SM, White AJ, Penninger JM, Lane PJL, Jenkinson EJ, Jenkinson WE, Anderson G. Developmentally regulated availability of RANKL and CD40 ligand reveals distinct mechanisms of fetal and adult cross-talk in the thymus medulla. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 189:5519-26. [PMID: 23152561 PMCID: PMC3605790 DOI: 10.4049/jimmunol.1201815] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
T cell tolerance in the thymus is a key step in shaping the developing T cell repertoire. Thymic medullary epithelial cells play multiple roles in this process, including negative selection of autoreactive thymocytes, influencing thymic dendritic cell positioning, and the generation of Foxp3(+) regulatory T cells. Previous studies show that medullary thymic epithelial cell (mTEC) development involves hemopoietic cross-talk, and numerous TNFR superfamily members have been implicated in this process. Whereas CD40 and RANK represent key examples, interplay between these receptors, and the individual cell types providing their ligands at both fetal and adult stages of thymus development, remain unclear. In this study, by analysis of the cellular sources of receptor activator for NF-κB ligand (RANKL) and CD40L during fetal and adult cross-talk in the mouse, we show that the innate immune cell system drives initial fetal mTEC development via expression of RANKL, but not CD40L. In contrast, cross-talk involving the adaptive immune system involves both RANKL and CD40L, with analysis of distinct subsets of intrathymic CD4(+) T cells revealing a differential contribution of CD40L by conventional, but not Foxp3(+) regulatory, T cells. We also provide evidence for a stepwise involvement of TNFRs in mTEC development, with CD40 upregulation induced by initial RANK signaling subsequently controlling proliferation within the mTEC compartment. Collectively, our findings show how multiple hemopoietic cell types regulate mTEC development through differential provision of RANKL/CD40L during ontogeny, revealing molecular differences in fetal and adult hemopoietic cross-talk. They also suggest a stepwise process of mTEC development, in which RANK is a master player in controlling the availability of other TNFR family members.
Collapse
Affiliation(s)
- Guillaume E. Desanti
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jennifer Cowan
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Song Baik
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Sonia M. Parnell
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Andrea J. White
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Josef M. Penninger
- Institute of Molecular Biotechnology, Austrian Academy of Science, Austria
| | - Peter J. L. Lane
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Eric J. Jenkinson
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - William E. Jenkinson
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Graham Anderson
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|