51
|
Nakajima A, Negishi N, Tsurui H, Kadowaki-Ohtsuji N, Maeda K, Nanno M, Yamaguchi Y, Shimizu N, Yagita H, Okumura K, Habu S. Commensal bacteria regulate thymic Aire expression. PLoS One 2014; 9:e105904. [PMID: 25157574 PMCID: PMC4144919 DOI: 10.1371/journal.pone.0105904] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/26/2014] [Indexed: 01/20/2023] Open
Abstract
Commensal bacteria in gastrointestinal tracts are reported to function as an environmental factor to regulate intestinal inflammation and immune responses. However, it remains largely unknown whether such bacterial function exerts any effect on other immune organs distant from the intestine. In this study, the influence of commensal bacteria in the thymus, where T cell lineages develop into mature type to form proper repertoires, was investigated using germ-free (GF) mice and Nod1-deficient mice lacking an intracellular recognition receptor for certain bacterial components, in which a commensal bacterial effect is predicted to be less. In both mice, there was no significant difference in the numbers and subset ratios of thymocytes. Interestingly, however, autoimmune regulator (Aire) expression in thymic epithelial cells (TECs), main components of the thymic microenvironment, was decreased in comparison to specific pathogen-free (SPF) mice and Nod1 wild-type (WT) mice, respectively. In vitro analysis using a fetal thymus organ culture (FTOC) system showed that Aire expression in TECs was increased in the presence of a bacterial component or a bacterial product. These results suggest that through their products, commensal bacteria have the potential to have some effect on epithelial cells of the thymus in tissues distant from the intestine where they are originally harbored.
Collapse
Affiliation(s)
- Akihito Nakajima
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Naoko Negishi
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Hiromichi Tsurui
- Department of Pathology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Naomi Kadowaki-Ohtsuji
- Department of Pathology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Keiko Maeda
- Atopy (Allergy) Research Center, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Masanobu Nanno
- Yakult Central Institute for Microbiological Research, Kunitachi, Tokyo, Japan
| | - Yoshitaka Yamaguchi
- Advanced Research Center for Genome Super Power, Keio University, Tsukuba, Ibaraki, Japan
| | - Nobuyoshi Shimizu
- Advanced Research Center for Genome Super Power, Keio University, Tsukuba, Ibaraki, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Ko Okumura
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
- Atopy (Allergy) Research Center, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Sonoko Habu
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
52
|
Yi Z, Stunz LL, Lin WW, Bishop GA. TRAF3 regulates homeostasis of CD8+ central memory T cells. PLoS One 2014; 9:e102120. [PMID: 25010048 PMCID: PMC4092107 DOI: 10.1371/journal.pone.0102120] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 06/06/2014] [Indexed: 11/23/2022] Open
Abstract
Our laboratory reported previously that TNF receptor associated factor 3 (TRAF3) is a positive regulator of TCR signaling and T cell function. In the current study, we present new findings that reveal differential roles for TRAF3 in the regulation of CD4+ and CD8+ T cells. In response to TCR stimulation in vitro, TRAF3 has greater impact in CD4+ T cells than in CD8+ T cells. However, T cell-specific TRAF3 deficient mice (CD4Cre TRAF3fl°x/fl°x; T-TRAF3−/−) have a greater number of CD4+CD44hi effector/memory T cells than littermate control (LMC) mice, possibly due to an inefficient suppressive effect of TRAF3 deficient Foxp3+ regulatory T cells. In contrast, CD8+CD44hiCD62Lhi central memory (Tcm) cells are markedly reduced in T-TRAF3−/− mice in comparison to LMC mice, although CD8+CD44hiCD62Ll°w effector memory T (Tem) cells and naïve T cells (CD8+CD44l°wCD62Lhi) do not show significant differences in number. Importantly, TRAF3-deficient Tcm cells exhibit defective homeostasis due to impaired IL-15 signaling. These results indicate that the involvement of TRAF3 in IL-15 mediated signaling to T cells plays a previously unappreciated and critical role in CD8+ Tcm cell regulation and maintenance.
Collapse
Affiliation(s)
- Zuoan Yi
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Laura L. Stunz
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Wai Wai Lin
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
- Graduate Immunology Program, University of Iowa, Iowa City, Iowa, United States of America
| | - Gail A. Bishop
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
- Graduate Immunology Program, University of Iowa, Iowa City, Iowa, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
- VA Medical Center, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
53
|
Xu X, Ge Q. Maturation and migration of murine CD4 single positive thymocytes and thymic emigrants. Comput Struct Biotechnol J 2014; 9:e201403003. [PMID: 24757506 PMCID: PMC3995209 DOI: 10.5936/csbj.201403003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/18/2014] [Accepted: 03/23/2014] [Indexed: 11/22/2022] Open
Abstract
T lymphopoiesis in the thymus was thought to be completed once they reach the single positive (SP) stage, when they are “fully mature” and wait to be exported at random or follow a “first in-first out” manner. Recently, accumulating evidence has revealed that newly generated SP thymocytes undergo further maturation in the thymic medulla before they follow a tightly regulated emigrating process to become recent thymic emigrants (RTEs). RTEs in the periphery then experience a post-thymic maturation and peripheral tolerance and eventually become licensed as mature naïve T cells. This review summarizes the recent progress in the late stage T cell development in and outside of the thymus. The regulation of this developmental process is also discussed.
Collapse
Affiliation(s)
- Xi Xu
- Key Laboratory of Medical Immunology, Ministry of Health. Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100191, P R China
| | - Qing Ge
- Key Laboratory of Medical Immunology, Ministry of Health. Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100191, P R China
| |
Collapse
|
54
|
Wolden-Kirk H, Rondas D, Bugliani M, Korf H, Van Lommel L, Brusgaard K, Christesen HT, Schuit F, Proost P, Masini M, Marchetti P, Eizirik DL, Overbergh L, Mathieu C. Discovery of molecular pathways mediating 1,25-dihydroxyvitamin D3 protection against cytokine-induced inflammation and damage of human and male mouse islets of Langerhans. Endocrinology 2014; 155:736-47. [PMID: 24424042 DOI: 10.1210/en.2013-1409] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Protection against insulitis and diabetes by active vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), in nonobese diabetic mice has until now mainly been attributed to its immunomodulatory effects, but also protective effects of this hormone on inflammation-induced β-cell death have been reported. The aim of this study was to clarify the molecular mechanisms by which 1,25(OH)2D3 contributes to β-cell protection against cytokine-induced β-cell dysfunction and death. Human and mouse islets were exposed to IL-1β and interferon-γ in the presence or absence of 1,25(OH)2D3. Effects on insulin secretion and β-cell survival were analyzed by glucose-stimulated insulin release and electron microscopy or Hoechst/propidium iodide staining, respectively. Gene expression profiles were assessed by Affymetrix microarrays. Nuclear factor-κB activity was tested, whereas effects on secreted chemokines/cytokines were confirmed by ELISA and migration studies. Cytokine exposure caused a significant increase in β-cell apoptosis, which was almost completely prevented by 1,25(OH)2D3. In addition, 1,25(OH)2D3 restored insulin secretion from cytokine-exposed islets. Microarray analysis of murine islets revealed that the expression of approximately 4000 genes was affected by cytokines after 6 and 24 hours (n = 4; >1.3-fold; P < .02), of which nearly 250 genes were modified by 1,25(OH)2D3. These genes belong to functional groups involved in immune response, chemotaxis, cell death, and pancreatic β-cell function/phenotype. In conclusion, these findings demonstrate a direct protective effect of 1,25(OH)2D3 against inflammation-induced β-cell dysfunction and death in human and murine islets, with, in particular, alterations in chemokine production by the islets. These effects may contribute to the beneficial effects of 1,25(OH)2D3 against the induction of autoimmune diabetes.
Collapse
Affiliation(s)
- H Wolden-Kirk
- Clinical and Experimental Endocrinology (H.W.-K., D.R., H.K., L.O., C.M.), University Hospital Gasthuisberg, Gene Expression Unit (L.V.L., F.S.), Department of Molecular and Cellular Medicine, Department of Microbiology and Immunology (P.P.), B-3000 Leuven, Belgium; Hans Christian Andersen Children's Hospital (H.W.-K., H.T.C.) and Department of Clinical Genetics (K.B., D.L.E.), Odense University Hospital, DK-5000, Odense, Denmark; Department of Endocrinology and Metabolism (M.B., P.M.), Metabolic Unit, and Department of General Pathology (M.M.), University of Pisa, Pisa, Italy; and Laboratory of Experimental Medicine (D.L.E.), Université Libre de Bruxelles, B-1070 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Gerondakis S, Fulford TS, Messina NL, Grumont RJ. NF-κB control of T cell development. Nat Immunol 2014; 15:15-25. [PMID: 24352326 DOI: 10.1038/ni.2785] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 11/12/2013] [Indexed: 12/12/2022]
Abstract
The NF-κB signal transduction pathway is best known as a major regulator of innate and adaptive immune responses, yet there is a growing appreciation of its importance in immune cell development, particularly of T lineage cells. In this Review, we discuss how the temporal regulation of NF-κB controls the stepwise differentiation and antigen-dependent selection of conventional and specialized subsets of T cells in response to T cell receptor and costimulatory, cytokine and growth factor signals.
Collapse
Affiliation(s)
- Steve Gerondakis
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Thomas S Fulford
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Nicole L Messina
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Raelene J Grumont
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| |
Collapse
|
56
|
Eshima K, Okabe M, Kajiura S, Noma H, Shinohara N, Iwabuchi K. Significant involvement of nuclear factor-κB-inducing kinase in proper differentiation of αβ and γδ T cells. Immunology 2014; 141:222-32. [PMID: 24117043 PMCID: PMC3904243 DOI: 10.1111/imm.12186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 01/07/2023] Open
Abstract
Nuclear factor-κB-inducing kinase (NIK) is known to play a critical role in maintaining proper immune function. This is exemplified in the spontaneous mutant mouse lacking functional NIK, alymphoplasia (aly), which is simultaneously immune-compromised and autoimmune-prone. To investigate the role of NIK in αβ T-cell repertoire formation, we analysed T-cell development in aly/aly mice bearing a transgenic T-cell receptor (TCR). Although there were no apparent abnormalities in the mature αβ T cells of non-transgenic aly/aly mice, the maturation efficiency of idiotype(high+) T cells in the TCR-transgenic mice was lower in aly/aly mice compared with those found in aly/+ mice, suggesting that the mature αβ T-cell repertoire could be altered by the absence of functional NIK. In one strain of TCR-transgenic aly/aly mice with a negatively selecting H-2 background, the proportion of CD8(low+) idiotype(high+) cells, which are thought to potentially represent the γδ lineage of T cells, was markedly decreased. When the γδ T cells in non-transgenic aly/aly mice were investigated, the proportion of γδ T cells in the peripheral organs of aly/aly mice was found to be one-half to one-fifth of those in aly/+ mice. Analyses of bone marrow chimera mice indicated that NIK in host cells, rather than in donor cells was important for generating a normal number of peripheral γδ T cells. Collectively, these results suggest that NIK could be involved in thymic positive selection of some αβ T cells and that NIK in non-haematopoietic cells is important for the optimal development and/or maintenance of γδ T cells.
Collapse
Affiliation(s)
- Koji Eshima
- Department of Immunology, Kitasato University School of Medicine, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
57
|
Roles for TNF-receptor associated factor 3 (TRAF3) in lymphocyte functions. Cytokine Growth Factor Rev 2013; 25:147-56. [PMID: 24433987 DOI: 10.1016/j.cytogfr.2013.12.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/15/2013] [Indexed: 12/27/2022]
Abstract
TRAF3 is an adapter protein that serves and regulates the functions of several types of receptors, located both inside the cell and at the plasma membrane. These include members of the TNF receptor superfamily (TNFR-SF), toll-like receptors (TLR), and cytokine receptors. It has become increasingly evident that the roles and functions of TRAF3 are highly context-dependent. TRAF3 can serve distinct roles for different receptors in the same cell, and also has highly cell-type-dependent functions. This review focuses upon the current state of knowledge regarding how TRAF3 regulates the biology and effector functions of B and T lymphocytes, two major cell types of the adaptive immune response in which TRAF3 has markedly distinct roles.
Collapse
|
58
|
Bayer AL, Pugliese A, Malek TR. The IL-2/IL-2R system: from basic science to therapeutic applications to enhance immune regulation. Immunol Res 2013; 57:197-209. [PMID: 24214027 PMCID: PMC3990437 DOI: 10.1007/s12026-013-8452-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
IL-2 plays a critical role in the normal function of the immune system. A trophic factor for lymphocytes, IL-2 is required for mounting and sustaining adaptive T cell responses; however, IL-2 is also critical for immune regulation via its effects on regulatory T cells (Treg cells). Over the years, we have contributed to the understanding of the biology of IL-2 and its signaling through the IL-2 receptor and helped define the key role played by IL-2 in Treg development and function. Our data show that Treg cells have a heightened sensitivity to IL-2, which may create a therapeutic window to promote immune regulation by selective stimulation of Treg cells. We are now developing new efforts to translate this knowledge to the clinical arena, through our focused interest in Type 1 diabetes as a prototypic autoimmune disease. Specifically, we aim at developing IL-2-based therapeutic regimens and incorporate means to enhance antigen-specific Treg responses, for improved and more selective regulation of islet autoimmunity. In parallel, we are pursuing studies in preclinical models of autoimmunity and transplantation to define critical factors for successful adoptive Treg therapy and develop clinically applicable therapeutic protocols.
Collapse
Affiliation(s)
- Allison L. Bayer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
| | - Alberto Pugliese
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
| | - Thomas R. Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
| |
Collapse
|
59
|
Maturation and emigration of single-positive thymocytes. Clin Dev Immunol 2013; 2013:282870. [PMID: 24187562 PMCID: PMC3804360 DOI: 10.1155/2013/282870] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/01/2013] [Indexed: 01/01/2023]
Abstract
T lymphopoiesis in the thymus was thought to be completed once it reaches the single positive (SP)
stage, a stage when T cells are “fully mature” and waiting to be exported at random or follow a “first-in-first-out” manner. Recent evidence, however, has revealed that the newly generated SP thymocytes undergo a multistage maturation program in the thymic medulla. Such maturation is followed by a tightly regulated emigration process and a further postthymic maturation of recent thymic emigrants (RTEs). This review summarizes recent progress in the late stage T cell development. The regulation of this developmental process is discussed.
Collapse
|
60
|
Murray SE. Cell-intrinsic role for NF-kappa B-inducing kinase in peripheral maintenance but not thymic development of Foxp3+ regulatory T cells in mice. PLoS One 2013; 8:e76216. [PMID: 24073289 PMCID: PMC3779168 DOI: 10.1371/journal.pone.0076216] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/21/2013] [Indexed: 11/18/2022] Open
Abstract
NF-κB inducing kinase (NIK, MAP3K14) is a key signaling molecule in non-canonical NF-κB activation, and NIK deficient mice have been instrumental in deciphering the immunologic role of this pathway. Global ablation of NIK prevents lymph node development, impairs thymic stromal development, and drastically reduces B cells. Despite altered thymic selection, T cell numbers are near normal in NIK deficient mice. The exception is CD4(+) regulatory T cells (Tregs), which are reduced in the thymus and periphery. Defects in thymic stroma are known to contribute to impaired Treg generation, but whether NIK also plays a cell intrinsic role in Tregs is unknown. Here, we compared intact mice with single and mixed BM chimeric mice to assess the intrinsic role of NIK in Treg generation and maintenance. We found that while NIK expression in stromal cells suffices for normal thymic Treg development, NIK is required cell-intrinsically to maintain peripheral Tregs. In addition, we unexpectedly discovered a cell-intrinsic role for NIK in memory phenotype conventional T cells that is masked in intact mice, but revealed in BM chimeras. These results demonstrate a novel role for NIK in peripheral regulatory and memory phenotype T cell homeostasis.
Collapse
Affiliation(s)
- Susan E. Murray
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
61
|
Akiyama T, Shinzawa M, Qin J, Akiyama N. Regulations of gene expression in medullary thymic epithelial cells required for preventing the onset of autoimmune diseases. Front Immunol 2013; 4:249. [PMID: 23986760 PMCID: PMC3752772 DOI: 10.3389/fimmu.2013.00249] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/09/2013] [Indexed: 11/13/2022] Open
Abstract
Elimination of potential self-reactive T cells in the thymus is crucial for preventing the onset of autoimmune diseases. Epithelial cell subsets localized in thymic medulla [medullary thymic epithelial cells (mTECs)] contribute to this process by supplying a wide range of self-antigens that are otherwise expressed in a tissue-specific manner (TSAs). Expression of some TSAs in mTECs is controlled by the autoimmune regulator (AIRE) protein, of which dysfunctional mutations are the causative factor of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED). In addition to the elimination of self-reactive T cells, recent studies indicated roles of mTECs in the development of Foxp3-positive regulatory T cells, which suppress autoimmunity and excess immune reactions in peripheral tissues. The TNF family cytokines, RANK ligand, CD40 ligand, and lymphotoxin were found to promote the differentiation of AIRE- and TSA-expressing mTECs. Furthermore, activation of NF-κB is essential for mTEC differentiation. In this mini-review, we focus on molecular mechanisms that regulate induction of AIRE and TSA expression and discuss possible contributions of these mechanisms to prevent the onset of autoimmune diseases.
Collapse
Affiliation(s)
- Taishin Akiyama
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | | | | | | |
Collapse
|
62
|
Shi Y, Zhu M. Medullary thymic epithelial cells, the indispensable player in central tolerance. SCIENCE CHINA. LIFE SCIENCES 2013; 56:392-8. [PMID: 23633070 DOI: 10.1007/s11427-013-4482-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 03/21/2013] [Indexed: 02/06/2023]
Abstract
Crosstalk between thymocytes and thymic epithelial cells is critical for T cell development and the establishment of central tolerance. Medullary thymic epithelial cells (mTECs) play important roles in the late stage of T cell development, especially negative selection and Treg generation. The function of mTECs is highly dependent on their characteristic features such as ectopic expression of peripheral tissue restricted antigens (TRAs) and their master regulator-autoimmune regulator (Aire), expression of various chemokines and cytokines. In this review, we summarize the current understanding of cellular and molecular mechanisms of mTEC development and its functions in T cell development and the establishment of central tolerance. The open questions in this field are also discussed. Understanding the function and underlying mechanisms of mTECs will contribute to the better control of autoimmune diseases and the improvement of immune reconstitution during aging or after infection, chemotherapy or radiotherapy.
Collapse
Affiliation(s)
- Yaoyao Shi
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | | |
Collapse
|
63
|
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
|
64
|
Abstract
It is widely accepted that Type 1 diabetes is a complex disease. Genetic predisposition and environmental factors favour the triggering of autoimmune responses against pancreatic β-cells, eventually leading to β-cell destruction. Over 40 susceptibility loci have been identified, many now mapped to known genes, largely supporting a dominant role for an immune-mediated pathogenesis. This role is also supported by the identification of several islet autoantigens and antigen-specific responses in patients with recent onset diabetes and subjects with pre-diabetes. Increasing evidence suggests certain viruses as a common environmental factor, together with diet and the gut microbiome. Inflammation and insulin resistance are emerging as additional cofactors, which might be interrelated with environmental factors. The heterogeneity of disease progression and clinical manifestations is likely a reflection of this multifactorial pathogenesis. So far, clinical trials have been mostly ineffective in delaying progression to overt diabetes in relatives at increased risk, or in reducing further loss of insulin secretion in patients with new-onset diabetes. This limited success may reflect, in part, our incomplete understanding of key pathogenic mechanisms, the lack of truly robust biomarkers of both disease activity and β-cell destruction, and the inability to assess the relative contributions of various pathogenic mechanisms at various time points during the course of the natural history of Type 1 diabetes. Emerging data and a re-evaluation of histopathological, immunological and metabolic findings suggest the hypothesis that unknown mechanisms of β-cell dysfunction may be present at diagnosis, and may contribute to the development of hyperglycaemia and clinical symptoms.
Collapse
Affiliation(s)
- A Pugliese
- Diabetes Research Institute, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
65
|
García-Lozano JR, Torres-Agrela B, Montes-Cano MA, Ortiz-Fernández L, Conde-Jaldón M, Teruel M, García A, Núñez-Roldán A, Martín J, González-Escribano MF. Association of the AIRE gene with susceptibility to rheumatoid arthritis in a European population: a case control study. Arthritis Res Ther 2013; 15:R11. [PMID: 23320549 PMCID: PMC3672784 DOI: 10.1186/ar4141] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 01/03/2013] [Accepted: 01/09/2013] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION AIRE is a transcriptional regulator playing a functional role in thymocyte education and negative selection by controlling the expression of peripheral antigens in the thymus. Recently, the AIRE gene was identified as a genetic risk factor for rheumatoid arthritis (RA) in genome wide association (GWA) studies performed in the Japanese population. According to the available data this association is restricted to the Asian population. However, different facts could influence the lack of association in Caucasian populations. The aim of this study was to further investigate the possible role of the AIRE gene in susceptibility to RA in a Caucasian population. METHODS A total of 472 Spanish Caucasian RA patients and 475 ethnically matched controls were included in the study. Three single-nucleotide polymorphisms (SNPs) (rs2776377, rs878081 and rs1055311) with a minor allele frequency>0.05 in the Caucasian population which were not included in the high-throughput platforms used in the GWA studies performed in susceptibility to RA, and two SNPs (rs2075876 and rs1800520) associated with RA in the Japanese population, were selected and genotyped using TaqMan assays. RESULTS No significant differences in the distribution of the alleles of rs2776377, rs2075876, rs1055311 and rs1800520 SNPs between RA patients and controls were observed. Nevertheless, the frequency of the C allele of rs878081 was significantly higher among RA patients (80.5% vs. 74.6% in the control group, pc=0.012, OR=1.41, 95%CI 1.13-1.75). Regarding the distribution of the rs878081 genotypes, a higher frequency of CC homozygous individuals was found in the RA patient group (65.56% vs. 56.47% in the control group, pc=0.013, OR=1.47, 95%CI 1.12-1.93). The in silico analysis predicted lower affinity to the binding-site of a motif of the transcription NF-κB family and lower transcription levels of AIRE gene for the rs878081C risk variant CONCLUSIONS Our findings suggest that the AIRE gene is associated with susceptibility to RA in the Spanish population. Probably, this association has not been detected in the European population in the GWA studies because the earliest high-throughput platforms did not include SNP suitable markers (e.g. rs878081).
Collapse
Affiliation(s)
- José-Raúl García-Lozano
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Avenida Manuel Siurot s/n, 41013-Sevilla, Spain
| | - Belén Torres-Agrela
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Avenida Manuel Siurot s/n, 41013-Sevilla, Spain
| | - Marco-Antonio Montes-Cano
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Avenida Manuel Siurot s/n, 41013-Sevilla, Spain
| | - Lourdes Ortiz-Fernández
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Avenida Manuel Siurot s/n, 41013-Sevilla, Spain
| | - Marta Conde-Jaldón
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Avenida Manuel Siurot s/n, 41013-Sevilla, Spain
| | - María Teruel
- Instituto de Parasitología y Biomedicina "López Neyra", CSIC, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento s/n, Armilla, 18100-Granada, Spain
| | - Alicia García
- Unidad de Reumatología, Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot s/n, 41013-Sevilla, Spain
| | - Antonio Núñez-Roldán
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Avenida Manuel Siurot s/n, 41013-Sevilla, Spain
| | - Javier Martín
- Instituto de Parasitología y Biomedicina "López Neyra", CSIC, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento s/n, Armilla, 18100-Granada, Spain
| | | |
Collapse
|
66
|
Perniola R. Expression of the autoimmune regulator gene and its relevance to the mechanisms of central and peripheral tolerance. Clin Dev Immunol 2012; 2012:207403. [PMID: 23125865 PMCID: PMC3485510 DOI: 10.1155/2012/207403] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 08/26/2012] [Accepted: 09/11/2012] [Indexed: 01/12/2023]
Abstract
The autoimmune polyendocrine syndrome type 1 (APS-1) is a monogenic disease due to pathogenic variants occurring in the autoimmune regulator (AIRE) gene. Its related protein, AIRE, activates the transcription of genes encoding for tissue-specific antigens (TsAgs) in a subset of medullary thymic epithelial cells: the presentation of TsAgs to the maturating thymocytes induces the apoptosis of the autoreactive clones and constitutes the main form of central tolerance. Dysregulation of thymic AIRE expression in genetically transmitted and acquired diseases other than APS-1 may contribute to further forms of autoimmunity. As AIRE and its murine homolog are also expressed in the secondary lymphoid organs, the extent and relevance of AIRE participation in the mechanisms of peripheral tolerance need to be thoroughly defined.
Collapse
Affiliation(s)
- Roberto Perniola
- Neonatal Intensive Care, Department of Pediatrics, V. Fazzi Regional Hospital, Piazza F. Muratore, 73100 Lecce, Italy.
| |
Collapse
|
67
|
Akiyama T, Shinzawa M, Akiyama N. TNF receptor family signaling in the development and functions of medullary thymic epithelial cells. Front Immunol 2012; 3:278. [PMID: 22969770 PMCID: PMC3432834 DOI: 10.3389/fimmu.2012.00278] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 08/16/2012] [Indexed: 11/25/2022] Open
Abstract
Thymic epithelial cells (TECs) provide the microenvironment required for the development of T cells in the thymus. A unique property of medullary thymic epithelial cells (mTECs) is their expression of a wide range of tissue-restricted self-antigens, critically regulated by the nuclear protein AIRE, which contributes to the selection of the self-tolerant T cell repertoire, thereby suppressing the onset of autoimmune diseases. The TNF receptor family (TNFRF) protein receptor activator of NF-κB (RANK), CD40 and lymphotoxin β receptor (LtβR) regulate the development and functions of mTECs. The engagement of these receptors with their specific ligands results in the activation of the NF-κB family of transcription factors. Two NF-κB activation pathways, the classical and non-classical pathways, promote the development of mature mTECs induced by these receptors. Consistently, TNF receptor-associated factor (TRAF6), the signal transducer of the classical pathway, and NF-κB inducing kinase (NIK), the signal transducer of the non-classical pathway, are essential for the development of mature mTECs. This review summarizes the current understanding of how the signaling by the TNF receptor family controls the development and functions of mTEC.
Collapse
Affiliation(s)
- Taishin Akiyama
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo Tokyo, Japan
| | | | | |
Collapse
|
68
|
Gerondakis S, Banerjee A, Grigoriadis G, Vasanthakumar A, Gugasyan R, Sidwell T, Grumont RJ. NF-κB subunit specificity in hemopoiesis. Immunol Rev 2012; 246:272-85. [PMID: 22435561 DOI: 10.1111/j.1600-065x.2011.01090.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although the diverse functions served by the nuclear factor-κB (NF-κB) pathway in virtually all cell types are typically employed to deal with stress responses, NF-κB transcription factors also play key roles in the development of hemopoietic cells. This review focuses on how NF-κB transcription factors control various aspects of thymic T-cell and myeloid cell differentiation that include its roles in hemopoietic precursors, conventional αβ T cells, CD4(+) regulatory T cells, natural killer T cells, γδ T cells, macrophages, and dendritic cells.
Collapse
|
69
|
Abstract
Somatic recombination of TCR genes in immature thymocytes results in some cells with useful TCR specificities, but also many with useless or potentially self-reactive specificities. Thus thymic selection mechanisms operate to shape the T-cell repertoire. Thymocytes that have a TCR with low affinity for self-peptide-MHC complexes are positively selected to further differentiate and function in adaptive immunity, whereas useless ones die by neglect. Clonal deletion and clonal diversion (Treg differentiation) are the major processes in the thymus that eliminate or control self-reactive T cells. Although these processes are thought to be efficient, they fail to control self-reactivity in all circumstances. Thus, peripheral tolerance processes exist wherein self-reactive T cells become functionally unresponsive (anergy) or are deleted after encountering self-antigens outside of the thymus. Recent advances in mechanistic studies of central and peripheral T-cell tolerance are promoting the development of therapeutic strategies to treat autoimmune disease and cancer and improve transplantation outcome.
Collapse
Affiliation(s)
- Yan Xing
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, 55455, USA
| | | |
Collapse
|
70
|
Barnaba V, Paroli M, Piconese S. The ambiguity in immunology. Front Immunol 2012; 3:18. [PMID: 22566903 PMCID: PMC3341998 DOI: 10.3389/fimmu.2012.00018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 02/02/2012] [Indexed: 01/07/2023] Open
Abstract
In the present article, we discuss the various ambiguous aspects of the immune system that render this complex biological network so highly flexible and able to defend the host from different external invaders. This ambiguity stems mainly from the property of the immune system to be both protective and harmful. Immunity cannot be fully protective without producing a certain degree of damage (immunopathology) to the host. The balance between protection and tissue damage is, therefore, critical for the establishment of immune homeostasis and protection. In this review, we will consider as ambiguous, various immunological tactics including: (a) the opposing functions driving immune responses, immune-regulation, and contra-regulation, as well as (b) the phenomenon of chronic immune activation as a result of a continuous cross-presentation of apoptotic T cells by dendritic cells. All these plans participate principally to maintain a state of chronic low-level inflammation during persisting infections, and ultimately to favor the species survival.
Collapse
Affiliation(s)
- Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma Rome, Italy
| | | | | |
Collapse
|
71
|
Dynamics of Lymphocyte Populations during Trypanosoma cruzi Infection: From Thymocyte Depletion to Differential Cell Expansion/Contraction in Peripheral Lymphoid Organs. J Trop Med 2012; 2012:747185. [PMID: 22505943 PMCID: PMC3306984 DOI: 10.1155/2012/747185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 10/17/2011] [Indexed: 01/05/2023] Open
Abstract
The comprehension of the immune responses in infectious diseases is crucial for developing novel therapeutic strategies. Here, we review current findings on the dynamics of lymphocyte subpopulations following experimental acute infection by Trypanosoma cruzi, the causative agent of Chagas disease. In the thymus, although the negative selection process of the T-cell repertoire remains operational, there is a massive thymocyte depletion and abnormal release of immature CD4+CD8+ cells to peripheral lymphoid organs, where they acquire an activated phenotype similar to activated effector or memory T cells. These cells apparently bypassed the negative selection process, and some of them are potentially autoimmune. In infected animals, an atrophy of mesenteric lymph nodes is also observed, in contrast with the lymphocyte expansion in spleen and subcutaneous lymph nodes, illustrating a complex and organ specific dynamics of lymphocyte subpopulations. Accordingly, T- and B-cell activation is seen in subcutaneous lymph nodes and spleen, but not in mesenteric lymph nodes. Lastly, although the function of peripheral CD4+CD8+ T-cell population remains to be defined in vivo, their presence may contribute to the immunopathological events found in both murine and human Chagas disease.
Collapse
|
72
|
Lovewell T, Tazi-Ahnini R. Models to explore the molecular function and regulation of AIRE. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2011. [DOI: 10.1016/j.ejmhg.2011.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
73
|
Teng F, Zhou Y, Jin R, Chen Y, Pei X, Liu Y, Dong J, Wang W, Pang X, Qian X, Chen WF, Zhang Y, Ge Q. The molecular signature underlying the thymic migration and maturation of TCRαβ+ CD4+ CD8 thymocytes. PLoS One 2011; 6:e25567. [PMID: 22022412 PMCID: PMC3192722 DOI: 10.1371/journal.pone.0025567] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 09/05/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND After positive selection, the newly generated single positive (SP) thymocytes migrate to the thymic medulla, where they undergo negative selection to eliminate autoreactive T cells and functional maturation to acquire immune competence and egress capability. METHODOLOGY/PRINCIPAL FINDINGS To elucidate the genetic program underlying this process, we analyzed changes in gene expression in four subsets of mouse TCRαβ(+)CD4(+)CD8(-) thymocytes (SP1 to SP4) representative of sequential stages in a previously defined differentiation program. A genetic signature of the migration of thymocytes was thus revealed. CCR7 and PlexinD1 are believed to be important for the medullary positioning of SP thymocytes. Intriguingly, their expression remains at low levels in the newly generated thymocytes, suggesting that the cortex-medulla migration may not occur until the SP2 stage. SP2 and SP3 cells gradually up-regulate transcripts involved in T cell functions and the Foxo1-KLF2-S1P(1) axis, but a number of immune function-associated genes are not highly expressed until cells reach the SP4 stage. Consistent with their critical role in thymic emigration, the expression of S1P(1) and CD62L are much enhanced in SP4 cells. CONCLUSIONS These results support at the molecular level that single positive thymocytes undergo a differentiation program and further demonstrate that SP4 is the stage at which thymocytes acquire the immunocompetence and the capability of emigration from the thymus.
Collapse
Affiliation(s)
- Fei Teng
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Yubin Zhou
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Rong Jin
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Yu Chen
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Xiaoyan Pei
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Yuanfeng Liu
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Jie Dong
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Wei Wang
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Xuewen Pang
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Xiaoping Qian
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Wei-Feng Chen
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Yu Zhang
- Department of Immunology, Peking University Health Science Center, Beijing, China
- * E-mail: (QG); (Y. Zhang)
| | - Qing Ge
- Department of Immunology, Peking University Health Science Center, Beijing, China
- * E-mail: (QG); (Y. Zhang)
| |
Collapse
|
74
|
Hofmann J, Mair F, Greter M, Schmidt-Supprian M, Becher B. NIK signaling in dendritic cells but not in T cells is required for the development of effector T cells and cell-mediated immune responses. J Exp Med 2011; 208:1917-29. [PMID: 21807870 PMCID: PMC3171087 DOI: 10.1084/jem.20110128] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 06/29/2011] [Indexed: 12/24/2022] Open
Abstract
The canonical NF-κB pathway is a driving force for virtually all aspects of inflammation. Conversely, the role of the noncanonical NF-κB pathway and its central mediator NF-κB-inducing kinase (NIK) remains poorly defined. NIK has been proposed to be involved in the formation of T(H)17 cells, and its absence in T(H) cells renders them incapable of inducing autoimmune responses, suggesting a T cell-intrinsic role for NIK. Upon systematic analysis of NIK function in cell-mediated immunity, we found that NIK signaling is dispensable within CD4(+) T cells but played a pivotal role in dendritic cells (DCs). We discovered that NIK signaling is required in DCs to deliver co-stimulatory signals to CD4(+) T cells and that DC-restricted expression of NIK is sufficient to restore T(H)1 and T(H)17 responses as well as cell-mediated immunity in NIK(-/-) mice. When CD4(+) T cells developed in the absence of NIK-sufficient DCs, they were rendered anergic. Reintroduction of NIK into DCs allowed developing NIK(-/-) CD4(+) T cells to become functional effector populations and restored the development of autoimmune disease. Therefore, our data suggest that a population of thymic DCs requires NIK to shape the formation of most αβ CD4(+) T effector lineages during early development.
Collapse
MESH Headings
- Animals
- Clonal Anergy/genetics
- Clonal Anergy/immunology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Immunity, Cellular/physiology
- Mice
- Mice, Knockout
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/immunology
- Protein Serine-Threonine Kinases/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Signal Transduction/immunology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
- NF-kappaB-Inducing Kinase
Collapse
Affiliation(s)
- Janin Hofmann
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Florian Mair
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Marc Schmidt-Supprian
- Molecular Immunology and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
75
|
Morrot A, Terra-Granado E, Pérez AR, Silva-Barbosa SD, Milićević NM, Farias-de-Oliveira DA, Berbert LR, De Meis J, Takiya CM, Beloscar J, Wang X, Kont V, Peterson P, Bottasso O, Savino W. Chagasic thymic atrophy does not affect negative selection but results in the export of activated CD4+CD8+ T cells in severe forms of human disease. PLoS Negl Trop Dis 2011; 5:e1268. [PMID: 21858238 PMCID: PMC3156684 DOI: 10.1371/journal.pntd.0001268] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 06/21/2011] [Indexed: 11/19/2022] Open
Abstract
Extrathymic CD4+CD8+ double-positive (DP) T cells are increased in some pathophysiological conditions, including infectious diseases. In the murine model of Chagas disease, it has been shown that the protozoan parasite Trypanosoma cruzi is able to target the thymus and induce alterations of the thymic microenvironment and the lymphoid compartment. In the acute phase, this results in a severe atrophy of the organ and early release of DP cells into the periphery. To date, the effect of the changes promoted by the parasite infection on thymic central tolerance has remained elusive. Herein we show that the intrathymic key elements that are necessary to promote the negative selection of thymocytes undergoing maturation during the thymopoiesis remains functional during the acute chagasic thymic atrophy. Intrathymic expression of the autoimmune regulator factor (Aire) and tissue-restricted antigen (TRA) genes is normal. In addition, the expression of the proapoptotic Bim protein in thymocytes was not changed, revealing that the parasite infection-induced thymus atrophy has no effect on these marker genes necessary to promote clonal deletion of T cells. In a chicken egg ovalbumin (OVA)-specific T-cell receptor (TCR) transgenic system, the administration of OVA peptide into infected mice with thymic atrophy promoted OVA-specific thymocyte apoptosis, further indicating normal negative selection process during the infection. Yet, although the intrathymic checkpoints necessary for thymic negative selection are present in the acute phase of Chagas disease, we found that the DP cells released into the periphery acquire an activated phenotype similar to what is described for activated effector or memory single-positive T cells. Most interestingly, we also demonstrate that increased percentages of peripheral blood subset of DP cells exhibiting an activated HLA-DR+ phenotype are associated with severe cardiac forms of human chronic Chagas disease. These cells may contribute to the immunopathological events seen in the Chagas disease. The thymus is a primary lymphoid organ that plays an important role on the development of the immune system and maturation of the T cell repertoire. During the normal life span, this organ undergoes involution during the aging and also in the presence of a wide variety of infectious diseases. It has been shown that the protozoan parasite Trypanosoma cruzi is able to target the thymus and induce alterations of the thymic microenvironment. In the acute phase, this results in a severe atrophy of the organ and early release of immature double-positive (DP) T cells into the periphery. The effect of the changes promoted by the parasite infection on thymic central tolerance has remained not clear. The present study shows that the intrathymic key elements that promote the negative selection of thymocytes during the thymopoiesis remains functional in the acute chagasic thymic atrophy. However, we found that the DP cells released into the periphery acquire an activated phenotype and its high frequency in the peripheral blood are associated with severe cardiac forms of human chronic Chagas disease.
Collapse
Affiliation(s)
- Alexandre Morrot
- Department of Immunology, Microbiology Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Abstract
In the thymus, developing T cells that react against self-antigens with high affinity are deleted in the process of negative selection. An essential component of this process is the display of self-antigens, including those whose expression are usually restricted to specific tissues, to developing T cells within the thymus. The Autoimmune Regulator (Aire) gene plays a crucial role in the expression of tissue specific self-antigens within the thymus, and disruption of Aire function results in spontaneous autoimmunity in both humans and mice. Recent advances have been made in our understanding of how Aire influences the expression of thousands of tissue-specific antigens in the thymus. Additional roles of Aire, including roles in chemokine and cytokine expression, have also been revealed. Factors important in the differentiation of Aire-expressing medullary thymic epithelial cells have been defined. Finally, the identity of antigen presenting cells in negative selection, including the role of medullary thymic epithelial cells in displaying tissue specific antigens to T cells, has also been clarified.
Collapse
Affiliation(s)
- Mark S. Anderson
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Maureen A. Su
- Inflammatory Diseases Institute and Department of Pediatrics, University of North Carolina, Chapel Hill, Chapel Hill, NC
| |
Collapse
|
77
|
Mouri Y, Yano M, Shinzawa M, Shimo Y, Hirota F, Nishikawa Y, Nii T, Kiyonari H, Abe T, Uehara H, Izumi K, Tamada K, Chen L, Penninger JM, Inoue JI, Akiyama T, Matsumoto M. Lymphotoxin signal promotes thymic organogenesis by eliciting RANK expression in the embryonic thymic stroma. THE JOURNAL OF IMMUNOLOGY 2011; 186:5047-57. [PMID: 21441458 DOI: 10.4049/jimmunol.1003533] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
It has recently become clear that signals mediated by members of the TNFR superfamily, including lymphotoxin-β receptor (LTβR), receptor activator for NF-κB (RANK), and CD40, play essential roles in organizing the integrity of medullary thymic epithelial cells (mTECs) required for the establishment of self-tolerance. However, details of the mechanism responsible for the unique and cooperative action of individual and multiple TNFR superfamily members during mTEC differentiation still remain enigmatic. In this study, we show that the LTβR signal upregulates expression of RANK in the thymic stroma, thereby promoting accessibility to the RANK ligand necessary for mTEC differentiation. Cooperation between the LTβR and RANK signals for optimal mTEC differentiation was underscored by the exaggerated defect of thymic organogenesis observed in mice doubly deficient for these signals. In contrast, we observed little cooperation between the LTβR and CD40 signals. Thus, the LTβR signal exhibits a novel and unique function in promoting RANK activity for mTEC organization, indicating a link between thymic organogenesis mediated by multiple cytokine signals and the control of autoimmunity.
Collapse
Affiliation(s)
- Yasuhiro Mouri
- Division of Molecular Immunology, Institute for Enzyme Research, The University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Pessach IM. The relationship of x-linked primary immune deficiencies and autoimmunity. Curr Allergy Asthma Rep 2010; 10:311-9. [PMID: 20571932 DOI: 10.1007/s11882-010-0127-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It is well-known that autoimmunity is significantly more prevalent in females. Growing evidence indicates that genes located on the X chromosome may play a role in autoimmunity and immune dysregulation, as also indicated by the frequent association of autoimmune phenomena in patients with X-linked primary immune deficiencies (PIDs). Hence, this group of genetic disorders is of particular interest to study PID-causing genes in the setting of more complex autoimmune disorders. This review focuses on the mechanisms leading to the autoimmune phenomena that are associated with the different X-linked PIDs, and on the intriguing interplay between immune dysregulation and immune deficiency in this unique setting.
Collapse
Affiliation(s)
- Itai M Pessach
- Division of Immunology, Children's Hospital, Harvard Medical School, One Blackfan Circle, Boston, MA 02115, USA.
| |
Collapse
|
79
|
Zhu M, Brown NK, Fu YX. Direct and indirect roles of the LTbetaR pathway in central tolerance induction. Trends Immunol 2010; 31:325-31. [PMID: 20675191 DOI: 10.1016/j.it.2010.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 06/19/2010] [Accepted: 06/22/2010] [Indexed: 12/30/2022]
Abstract
Medullary thymic epithelial cells (mTECs) play a critical role in thymic negative selection of autoreactive thymocytes, especially for thymocytes specific for peripheral tissue-restricted self-antigens (TRA). Deficiency in lymphotoxin b receptor (LTbetaR) is associated with peripheral tissue inflammation, but whether this is caused by defective negative selection has been unclear; the significance of the LTbetaR pathway for negative selection is evident in some models but not others. Here, we revisit the data and clarify the role of LTbetaR in mTEC development and function and thymic TRA expression. These processes are discussed as potential mechanisms for LTbetaR-mediated control of negative selection.
Collapse
Affiliation(s)
- Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | | | | |
Collapse
|
80
|
Autoimmune Regulator Gene (Aire) is Expressed in Lymph Node Fibroblastic Reticular Cell, BLS4. ACTA ACUST UNITED AC 2010. [DOI: 10.5352/jls.2010.20.5.670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
81
|
Zhu M, Fu Y. The complicated role of NF-kappaB in T-cell selection. Cell Mol Immunol 2010; 7:89-93. [PMID: 20190822 PMCID: PMC4001888 DOI: 10.1038/cmi.2009.112] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 12/02/2009] [Accepted: 12/05/2009] [Indexed: 02/08/2023] Open
Abstract
The nuclear factor (NF)-kappaB transcription factor family plays important roles in the immune system. Aberrant NF-kappaB signaling is frequently associated with inflammation and autoimmune diseases but the underlying mechanisms are not fully understood. Recent studies show that NF-kappaB plays a critical role in T-cell central tolerance. Two NF-kappaB signaling pathways have been identified: the canonical pathway and the alternative pathway. In the establishment of T-cell central tolerance, the alternative pathway appears to be the key signaling component in thymic stromal cells for their development and function, while the canonical pathway exerts its function more in autonomous T-cell selection. This review intends to summarize the current understanding of the role of NF-kappaB in establishing T-cell central tolerance and highlight unsolved intriguing questions for future work.
Collapse
Affiliation(s)
- Mingzhao Zhu
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing, China.
| | | |
Collapse
|
82
|
Transcriptional regulation of thymus organogenesis and thymic epithelial cell differentiation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 92:103-20. [PMID: 20800818 DOI: 10.1016/s1877-1173(10)92005-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Transcriptional regulatory networks are the central regulatory mechanisms that control organ identity, patterning, and differentiation. In the case of the thymus, several key transcription factors have been identified that are critical for various aspects of thymus organogenesis and thymic epithelial cell (TEC) differentiation. The thymus forms from the third pharyngeal pouch endoderm during embryogenesis. Organ development progresses from initial thymus cell fate specification, through multiple stages of TEC differentiation and cortical (cTEC) and medullary (mTEC) formation. Transcription factors have been identified for each of these stages: a Hoxa3-dependent cascade at initial fate specification, Foxn1 for early (and later) TEC differentiation, and NF-kappaB for mTEC differentiation. As important as these factors are, their interrelationships are not understood, and many more transcription factors are likely required for complete thymus organogenesis to occur. In this chapter, we review the literature on these known genes, as well as identify gaps in our knowledge for future studies.
Collapse
|
83
|
Abstract
This article focuses on the functions of NF-kappaB that vitally impact lymphocytes and thus adaptive immunity. NF-kappaB has long been known to be essential for many of the responses of mature lymphocytes to invading pathogens. In addition, NF-kappaB has important functions in shaping the immune system so it is able to generate adaptive responses to pathogens. In both contexts, NF-kappaB executes critical cell-autonomous functions within lymphocytes as well as within supportive cells, such as antigen-presenting cells or epithelial cells. It is these aspects of NF-kappaB's physiologic impact that we address in this article.
Collapse
|
84
|
Martin-Pagola A, Pileggi A, Zahr E, Vendrame F, Damaris Molano R, Snowhite I, Ricordi C, Eisenbarth GS, Nakayama M, Pugliese A. Insulin2 gene (Ins2) transcription by NOD bone marrow-derived cells does not influence autoimmune diabetes development in NOD-Ins2 knockout mice. Scand J Immunol 2009; 70:439-46. [PMID: 19874548 DOI: 10.1111/j.1365-3083.2009.02316.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Insulin is a critical autoantigen for the development of autoimmune diabetes in non-obese diabetic (NOD) mice. About 80% of NOD females and 30-40% of NOD males develop diabetes. However, Insulin2 (Ins2) knockout NOD mice develop autoimmune diabetes with complete penetrance in both sexes, at an earlier age, and have stronger autoimmune responses to insulin. The severe diabetes phenotype observed in NOD-Ins2-/- mice suggests that lack of Ins2 expression in the thymus may compromise immunological tolerance to insulin. Insulin is a prototypical tissue specific antigen (TSA) for which tolerance is dependent on expression in thymus and peripheral lymphoid tissues. TSA are naturally expressed by medullary thymic epithelial cells (mTEC), stromal cells in peripheral lymphoid tissues and bone marrow (BM)-derived cells, mainly CD11c(+) dendritic cells. The natural expression of TSA by mTEC and stromal cells has been shown to contribute to self-tolerance. However, it is unclear whether this also applies to BM-derived cells naturally expressing TSA. To address this question, we created BM chimeras and investigated whether reintroducing Ins2 expression solely by NOD BM-derived cells delays diabetes development in NOD-Ins2-/- mice. On follow-up, NOD-Ins2-/- mice receiving Ins2-expressing NOD BM cells developed diabetes at similar rates of those receiving NOD-Ins2-/- BM cells. Diabetes developed in 64% of NOD recipients transplanted with NOD BM and in 47% of NOD mice transplanted with NOD-Ins2-/- BM (P = ns). Thus, NOD-Ins2-/- BM did not worsen diabetes in NOD recipients and Ins2 expression by NOD BM-derived cells did not delay diabetes development in NOD-Ins2-/- mice.
Collapse
Affiliation(s)
- A Martin-Pagola
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Pessach IM, Notarangelo LD. X-linked primary immunodeficiencies as a bridge to better understanding X-chromosome related autoimmunity. J Autoimmun 2009; 33:17-24. [PMID: 19361956 DOI: 10.1016/j.jaut.2009.03.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 03/03/2009] [Accepted: 03/11/2009] [Indexed: 12/30/2022]
Abstract
Recent studies indicate that genes located on the X-chromosome play a major and unique role in autoimmunity. The fact that most X-linked primary immune deficiencies carry significant autoimmune manifestations greatly supports this notion. Autoimmunity and immune deficiency have been considered two opposite extremes resulting from immune dysregulation and failure of immune development and/or function, respectively. Growing evidence has been accumulating to indicate that autoimmune phenomena occur in patients suffering from primary immune deficiency (PID), and the molecular and cellular mechanisms that interconnect these conditions are being unraveled. The study of rare single-gene disorders associated with significant autoimmunity may shed light on the pathophysiology of more complex multifactorial and polygenic autoimmune disorders. In this regard, primary immunodeficiencies represent unique "experiments of Nature" that illustrate the critical role played by single-gene products in the development, function and homeostasis of the immune system. In this review we will focus on the clinical features and on the cellular and molecular pathophysiology of the known X-linked PID in which autoimmune manifestations are more common, in the attempt to understand what single-gene defects can teach us on the role that key immune pathways and cellular processes may play to prevent autoimmunity.
Collapse
Affiliation(s)
- Itai M Pessach
- Division of Immunology, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
86
|
Fletcher AL, Seach N, Reiseger JJ, Lowen TE, Hammett MV, Scott HS, Boyd RL. Reduced thymic Aire expression and abnormal NF-kappa B2 signaling in a model of systemic autoimmunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:2690-9. [PMID: 19234163 DOI: 10.4049/jimmunol.0801752] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
The thymic stromal niche normally directs the production and export of a self-tolerant T cell repertoire. Many models of spontaneous autoimmunity, however, develop thymic architectural abnormalities before disease onset. Although this is suspected to affect central tolerance induction, creating an autoimmune predisposition, in-depth analysis of the microenvironment within these thymi is lacking, such that the mechanisms and likely direct effects on the T cell repertoire are unknown or speculative. Here we show that NZB mice, the first described model for systemic autoimmunity, demonstrate a complex thymic phenotype, including a lack of the autoimmune regulator (Aire), early defects in thymic epithelial cell (TEC) expansion, and evidence for altered NF-kappaB2 signaling. Analysis of medullary TEC revealed a numerical loss of the Aire-expressing MHC class II(high) (mTEC-high) subset as well reduced Aire protein and mRNA per cell. RelB expression was also reduced, while chemokines CCL19 and CCL21 were increased. Unexpectedly, the proportion of cortex and medulla in the NZB mice was normal from 36 wk, despite worsening architectural abnormalities. These data show that the NZB defect is more complex than previously appreciated, segregating into early numerical TEC deficiencies that correct with age, late degeneration of the niche architecture that does not affect TEC number, and a persistent reduction in Aire and RelB expression per cell acquired upon mTEC-high differentiation.
Collapse
Affiliation(s)
- Anne L Fletcher
- Immune Regeneration Laboratory, Monash University, Clayton, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
87
|
Cell Defence and Survival. GUIDE TO SIGNAL PATHWAYS IN IMMUNE CELLS 2009. [PMCID: PMC7123614 DOI: 10.1007/978-1-60327-538-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Central to immune defence mechanisms is the role of transcription factor nuclear factor kappa B (NF-kB). This is a complex biochemical topic with ever more controls revealed. NF-kB determines the production of proinflammatory cytokines and chemokines. Pharmacologists step in with possible means of control. Other systems involved in defence include the cyclooxygenase 2 (Cox-2) enzyme and perioxisome proliferator-activated receptors. Insulin receptor activation needs to be seen in context. The mTOR system directs uptake of nutrients by cells. mTOR is suppressed by rapamycin, whose usage is now quite considerable in the control of transplant rejection.
Collapse
|
88
|
Peterson P, Org T, Rebane A. Transcriptional regulation by AIRE: molecular mechanisms of central tolerance. Nat Rev Immunol 2008; 8:948-57. [PMID: 19008896 PMCID: PMC2785478 DOI: 10.1038/nri2450] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The negative selection of T cells in the thymus is necessary for the maintenance of self tolerance. Medullary thymic epithelial cells have a key function in this process as they express a large number of tissue-specific self antigens that are presented to developing T cells. Mutations in the autoimmune regulator (AIRE) protein cause a breakdown of central tolerance that is associated with decreased expression of self antigens in the thymus. In this Review, we discuss the role of AIRE in the thymus and recent advances in our understanding of how AIRE might function at the molecular level to regulate gene expression.
Collapse
Affiliation(s)
- Pärt Peterson
- Institute of General and Molecular Pathology, University of Tartu, Tartu 5O411, Estonia.
| | | | | |
Collapse
|
89
|
Akiyama T, Shimo Y, Yanai H, Qin J, Ohshima D, Maruyama Y, Asaumi Y, Kitazawa J, Takayanagi H, Penninger JM, Matsumoto M, Nitta T, Takahama Y, Inoue JI. The tumor necrosis factor family receptors RANK and CD40 cooperatively establish the thymic medullary microenvironment and self-tolerance. Immunity 2008; 29:423-37. [PMID: 18799149 DOI: 10.1016/j.immuni.2008.06.015] [Citation(s) in RCA: 377] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 04/09/2008] [Accepted: 06/20/2008] [Indexed: 12/30/2022]
Abstract
Medullary thymic epithelial cells (mTECs) establish T cell self-tolerance through the expression of autoimmune regulator (Aire) and peripheral tissue-specific self-antigens. However, signals underlying mTEC development remain largely unclear. Here, we demonstrate crucial regulation of mTEC development by receptor activator of NF-kappaB (RANK) and CD40 signals. Whereas only RANK signaling was essential for mTEC development during embryogenesis, in postnatal mice, cooperation between CD40 and RANK signals was required for mTEC development to successfully establish the medullary microenvironment. Ligation of RANK or CD40 on fetal thymic stroma in vitro induced mTEC development in a tumor necrosis factor-associated factor 6 (TRAF6)-, NF-kappaB inducing kinase (NIK)-, and IkappaB kinase beta (IKKbeta)-dependent manner. These results show that developmental-stage-dependent cooperation between RANK and CD40 promotes mTEC development, thereby establishing self-tolerance.
Collapse
Affiliation(s)
- Taishin Akiyama
- Division of Cellular and Molecular biology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokane-dai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Abstract
Three papers in this issue of Immunity (Akiyama et al., 2008; Hikosaka et al., 2008; Irla et al., 2008) together reveal coordinating roles for autoreactive T cells and TNF receptor superfamily members in the development of medullary thymic epithelial cells.
Collapse
Affiliation(s)
- Mingzhao Zhu
- The Department of Pathology and Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
91
|
Ferguson BJ, Cooke A, Peterson P, Rich T. Death in the AIRE. Trends Immunol 2008; 29:306-12. [PMID: 18515183 DOI: 10.1016/j.it.2008.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 03/14/2008] [Accepted: 03/14/2008] [Indexed: 12/17/2022]
Abstract
When thymic epithelia begin to synthesize peripheral tissue antigens such as insulin, we are seeing the result of autoimmune regulator (AIRE) activity and the workings of central tolerance. AIRE is an extraordinary protein that repatterns the transcriptome of medullary thymic epithelia (mTECs) to produce a stroma decorated with peripheral self-peptides. These peptidic arrays are used to purge self-reactive T cells, thereby averting autoimmunity. We now propose that an inherently cytotoxic event such as global chromatin modification paves the way for AIRE action. This injury stimulus might impose temporal restrictions for the T-cell education process and is endured, at least transiently, by the unique cellular environment provided by the medullary thymic epithelia.
Collapse
Affiliation(s)
- Brian J Ferguson
- Department of Pathology, Divisions of Immunology and Cellular Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | | | | | | |
Collapse
|
92
|
Carlsén M, Cilio CM. Evidence for de novo expression of thymic insulin by peripheral bone marrow-derived cells. Scand J Immunol 2008; 68:67-74. [PMID: 18482204 DOI: 10.1111/j.1365-3083.2008.02121.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Thymic expression of insulin has been suggested to play a major role in negative selection of autoreactive T cells and tolerance induction against pancreatic beta cells. Furthermore, the expression of insulin in peripheral antigen-presenting cells (APC) has been clearly demonstrated but whether thymic negative selection and tolerance induction also depends on peripheral influx of self-antigens (Ag) remains to be conclusively demonstrated. In this study, we wanted to test whether peripheral influx of insulin expressing cells might contribute to negative selection. In order to address this question, we used mice deficient in the Ins1 and Ins2 genes. Embryonic thymi either deficient in both insulin genes or expressing Ins2 were dissected and transplanted under the kidney capsule of athymic nude mice recipients. After indicated time points, grafted thymi were removed and analysed for insulin re-expression and for the emergence of autoreactive T cells. The analysis revealed a re-expression of Ins2 in grafted insulin deficient thymi suggesting that self-Ag expression in the thymus is not only intrinsically regulated but peripheral influx of APC capable of expressing insulin might contribute to thymic selection and tolerance induction.
Collapse
Affiliation(s)
- M Carlsén
- Cellular Autoimmunity Unit, Department of Clinical Sciences, Clinical Research Center, Malmö University Hospital, Lund University, Malmö, Sweden
| | | |
Collapse
|
93
|
Of Omenn and mice. Trends Immunol 2008; 29:133-40. [PMID: 18255337 DOI: 10.1016/j.it.2007.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 11/29/2007] [Accepted: 12/03/2007] [Indexed: 11/22/2022]
Abstract
Omenn syndrome (OS) is a peculiar immunodeficiency in which profound T and B cell defects are associated with severe autoimmune manifestations. Although the molecular and biochemical bases of OS have been elucidated, the mechanisms leading to T cell infiltration of peripheral tissues such as skin and gut still remain unsolved. Two murine models with hypomorphic mutations in rag genes reproducing OS features and a murine model of lymphopenia-derived autoimmunity with similar immunopathology were recently described. The aim of this review is to integrate clues regarding the roles of impaired thymic development and lymphopenia into the pathogenesis of autoimmunity.
Collapse
|
94
|
Tucker E, O'Donnell K, Fuchsberger M, Hilton AA, Metcalf D, Greig K, Sims NA, Quinn JM, Alexander WS, Hilton DJ, Kile BT, Tarlinton DM, Starr R. A novel mutation in the Nfkb2 gene generates an NF-kappa B2 "super repressor". THE JOURNAL OF IMMUNOLOGY 2008; 179:7514-22. [PMID: 18025196 DOI: 10.4049/jimmunol.179.11.7514] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The noncanonical NF-kappaB pathway regulates the development and function of multiple organs and cell lineages. We have generated mice harboring a novel mutation in Nfkb2 that prevents the processing of the inhibitory precursor, p100, into the active subunit, p52. Mutant mice express a complex phenotype with abnormalities in a variety of tissues, and with a spectrum that is more severe than in mice carrying a targeted deletion of Nfkb2. Signaling through the noncanonical pathway is ablated due to the absence of p52, resulting in disorganized splenic architecture and disrupted B cell development. The inhibitory precursor form of NF-kappaB2 interacts with RelA, preventing activation of RelA dimers in response to both canonical and noncanonical stimuli, which in combination with p52 deficiency, results in defective lymph node formation and bone homeostasis. These findings demonstrate a key role for NF-kappaB2 in the regulation of RelA activation and suggest overlap in the function of NF-kappaB members in canonical and noncanonical pathway signaling.
Collapse
Affiliation(s)
- Elena Tucker
- St Vincent's Institute, Fitzroy, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Ferguson BJ, Alexander C, Rossi SW, Liiv I, Rebane A, Worth CL, Wong J, Laan M, Peterson P, Jenkinson EJ, Anderson G, Scott HS, Cooke A, Rich T. AIRE's CARD revealed, a new structure for central tolerance provokes transcriptional plasticity. J Biol Chem 2008; 283:1723-1731. [PMID: 17974569 DOI: 10.1074/jbc.m707211200] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
Developing T cells encounter peripheral self-antigens in the thymus in order to delete autoreactive clones. It is now known that the autoimmune regulator protein (AIRE), which is expressed in thymic medullary epithelial cells, plays a key role in regulating the thymic transcription of these peripheral tissue-specific antigens. Mutations in the AIRE gene are associated with a severe multiorgan autoimmune syndrome (APECED), and autoimmune reactivities are manifest in AIRE-deficient mice. Functional AIRE protein is expressed as distinct nuclear puncta, although no structural basis existed to explain their relevance to disease. In addressing the cell biologic basis for APECED, we made the unexpected discovery that an AIRE mutation hot spot lies in a caspase recruitment domain. Combined homology modeling and in vitro data now show how APECED mutations influence the activity of this transcriptional regulator. We also provide novel in vivo evidence for AIRE's association with a global transcription cofactor, which may underlie AIRE's focal, genome-wide, alteration of the transcriptome.
Collapse
Affiliation(s)
- Brian J Ferguson
- Department of Pathology, Divisions of Immunology and Cellular Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Clare Alexander
- Department of Pathology, Divisions of Immunology and Cellular Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Simona W Rossi
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Ingrid Liiv
- Molecular Pathology, University of Tartu, Biomedicum, 50411 Tartu, Estonia
| | - Ana Rebane
- Molecular Pathology, University of Tartu, Biomedicum, 50411 Tartu, Estonia
| | - Catherine L Worth
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, United Kingdom
| | - Joyce Wong
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, United Kingdom
| | - Martti Laan
- Molecular Pathology, University of Tartu, Biomedicum, 50411 Tartu, Estonia
| | - Pärt Peterson
- Molecular Pathology, University of Tartu, Biomedicum, 50411 Tartu, Estonia
| | - Eric J Jenkinson
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Graham Anderson
- Medical Research Council Centre for Immune Regulation, Institute for Biomedical Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Hamish S Scott
- Walter and Eliza Hall Institute of Medical Research, 3050 Melbourne, Australia
| | - Anne Cooke
- Department of Pathology, Divisions of Immunology and Cellular Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Tina Rich
- Department of Pathology, Divisions of Immunology and Cellular Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom.
| |
Collapse
|
96
|
Chen J, Yang W, Yu C, Li Y. Autoimmune regulator initiates the expression of promiscuous genes in thymic epithelial cells. Immunol Invest 2008; 37:203-14. [PMID: 18389440 DOI: 10.1080/08820130801967841] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The expression of peripheral antigens in the thymus, known as promiscuous gene expression, has been implicated in T cell tolerance and autoimmunity. Here we identified thymic epithelial cells (TECs) as the main cell type that expresses a diverse range of tissue-restricted antigens (TRAs). The TECs of a common autoimmune (non-obese diabetic [NOD]) mouse model express much lower levels of an autoimmune regulator (Aire) and TRAs than normal (Balb/c) TECs. Transfection of an Aire plasmid led to increased levels of TRA expression in cultured TECs from Balb/c and NOD mice; an increase that was enhanced by the presence of thymocytes. These data show that Aire initiates promiscuous gene expression in TECs, and that this function might be under thymocyte control.
Collapse
Affiliation(s)
- Jibing Chen
- Department of Immunology, Norman Bethune Medical College of JiLin University, Chang Chun, China
| | | | | | | |
Collapse
|
97
|
Nitta T, Murata S, Ueno T, Tanaka K, Takahama Y. Thymic microenvironments for T-cell repertoire formation. Adv Immunol 2008; 99:59-94. [PMID: 19117532 DOI: 10.1016/s0065-2776(08)00603-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Functionally competent immune system includes a functionally competent T-cell repertoire that is reactive to foreign antigens but is tolerant to self-antigens. The repertoire of T cells is primarily formed in the thymus through positive and negative selection of developing thymocytes. Immature thymocytes that undergo V(D)J recombination of T-cell antigen receptor (TCR) genes and that express the virgin repertoire of TCRs are generated in thymic cortex. The recent discovery of thymoproteasomes, a molecular complex specifically expressed in cortical thymic epithelial cells (cTEC), has revealed a unique role of cTEC in cuing the further development of immature thymocytes in thymic cortex, possibly by displaying unique self-peptides that induce positive selection. Cortical thymocytes that receive TCR-mediated positive selection signals are destined to survive for further differentiation and are induced to express CCR7, a chemokine receptor. Being attracted to CCR7 ligands expressed by medullary thymic epithelial cells (mTEC), CCR7-expressing positively selected thymocytes relocate to thymic medulla. The medullary microenvironment displays another set of unique self-peptides for trimming positively selected T-cell repertoire to establish self-tolerance, via promiscuous expression of tissue-specific antigens by mTEC and efficient antigen presentation by dendritic cells. Recent results demonstrate that tumor necrosis factor (TNF) superfamily ligands, including receptor activating NF-kappaB ligand (RANKL), CD40L, and lymphotoxin, are produced by positively selected thymocytes and pivotally regulate mTEC development and thymic medulla formation.
Collapse
Affiliation(s)
- Takeshi Nitta
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | | | |
Collapse
|
98
|
Zhu M, Chin RK, Tumanov AV, Liu X, Fu YX. Lymphotoxin β Receptor Is Required for the Migration and Selection of Autoreactive T Cells in Thymic Medulla. THE JOURNAL OF IMMUNOLOGY 2007; 179:8069-75. [DOI: 10.4049/jimmunol.179.12.8069] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
99
|
Li J, Li Y, Yao JY, Jin R, Zhu MZ, Qian XP, Zhang J, Fu YX, Wu L, Zhang Y, Chen WF. Developmental pathway of CD4+CD8- medullary thymocytes during mouse ontogeny and its defect in Aire-/- mice. Proc Natl Acad Sci U S A 2007; 104:18175-80. [PMID: 17984055 PMCID: PMC2084316 DOI: 10.1073/pnas.0708884104] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Indexed: 01/19/2023] Open
Abstract
The newly generated single-positive (SP) thymocytes undergo further maturation in the thymic medulla before their emigration to the periphery. The present study was undertaken to validate a developmental program we proposed for CD4SP medullary thymocytes and to explore the mechanisms regulating this process. During mouse ontogeny, the emergence of different subsets of CD4SP thymocytes followed a strict temporal order from SP1 to SP4. Parallel to the transition in surface phenotype, a steady increase in function was observed. As further evidence, purified SP1 cells were able to sequentially give rise to SP2, SP3, and SP4 cells in intrathymic adoptive transfer and in culture. Notably, the development of CD4SP cells in the medulla seemed to be critically dependent on a functionally intact medullary epithelial cell compartment because Relb and Aire deficiency were found to cause severe blockage at the transition from SP3 to SP4. Taken together, this work establishes an ontogenetically and functionally relevant maturation program for CD4SP thymocytes. Precise dissection of this program should facilitate further inquiry into the molecular mechanisms governing normal thymocyte development and its disturbance in pathological conditions.
Collapse
Affiliation(s)
- Juan Li
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| | - Yan Li
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| | - Jin-Yan Yao
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| | - Rong Jin
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| | - Ming-Zhao Zhu
- Department of Pathology, University of Chicago, 5841 South Maryland Avenue, Chicago, IL 60637; and
| | - Xiao-Ping Qian
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| | - Jun Zhang
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| | - Yang-Xin Fu
- Department of Pathology, University of Chicago, 5841 South Maryland Avenue, Chicago, IL 60637; and
| | - Li Wu
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3050, Australia
| | - Yu Zhang
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| | - Wei-Feng Chen
- *Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| |
Collapse
|
100
|
Venanzi ES, Gray DHD, Benoist C, Mathis D. Lymphotoxin pathway and Aire influences on thymic medullary epithelial cells are unconnected. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:5693-700. [PMID: 17947641 DOI: 10.4049/jimmunol.179.9.5693] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The lymphotoxin pathway is critical for the development and maintenance of peripheral lymphoid organs. Mice with deficiencies in members of this pathway lack lymph nodes and Peyer's patches and have abnormal spleen architecture. These animals also develop autoantibodies to and lymphocytic infiltrates of multiple organs, provoking speculation that the lymphotoxin pathway may play a role in central tolerance induction. Indeed, a series of reports has claimed that lymphotoxin signals control the expression of Aire, a transcriptional regulator that is expressed in medullary epithelial cells of the thymus, mediates ectopic transcription of genes encoding a variety of peripheral tissue Ags, and promotes clonal deletion of self-reactive thymocytes. However, one report argued that lymphotoxin signals regulate the composition and organization of the thymus, particularly of the medullary epithelial compartment. Herein, we resolve this controversy in favor of the latter view. The expression and function of Aire were unaffected in medullary epithelial cells of mice lacking either lymphotoxin beta receptor or the lymphotoxin alpha-chain, and there was minimal overlap between the sets of genes controlled by Aire and lymphotoxin. Instead, both knockout lines showed abnormal medullary epithelial cell organization, and the line lacking the beta receptor had significantly fewer medullary epithelial cells. In short, the lymphotoxin pathway drives the developmental rather than selectional properties of thymic stromal cells.
Collapse
Affiliation(s)
- Emily S Venanzi
- Department of Medicine, Section on Immunology and Immunogenetics, Joslin Diabetes Center, Brigham and Women's Hospital, Boston, MA 02215, USA
| | | | | | | |
Collapse
|