Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
2.
Clin Transl Med ; 11(6): e427, 2021 06.
Article in English | MEDLINE | ID: mdl-34185416

ABSTRACT

Rhinovirus (RV) infections are associated with asthma exacerbations. MicroRNA-146a and microRNA-146b (miR-146a/b) are anti-inflammatory miRNAs that suppress signaling through the nuclear factor kappa B (NF-κB) pathway and inhibit pro-inflammatory chemokine production in primary human bronchial epithelial cells (HBECs). In the current study, we aimed to explore whether miR-146a/b could regulate cellular responses to RVs in HBECs and airways during RV-induced asthma exacerbation. We demonstrated that expression of miR-146a/b and pro-inflammatory chemokines was increased in HBECs and mouse airways during RV infection. However, transfection with cell-penetrating peptide (CPP)-miR-146a nanocomplexes before infection with RV significantly reduced the expression of the pro-inflammatory chemokines CCL5, IL-8 and CXCL1, increased interferon-λ production, and attenuated infection with the green fluorescent protein (GFP)-expressing RV-A16 in HBECs. Concordantly, compared to wild-type (wt) mice, Mir146a/b-/- mice exhibited more severe airway neutrophilia and increased T helper (Th)1 and Th17 cell infiltration in response to RV-A1b infection and a stronger Th17 response with a less prominent Th2 response in house dust mite extract (HDM)-induced allergic airway inflammation and RV-induced exacerbation models. Interestingly, intranasal administration of CPP-miR-146a nanocomplexes reduced HDM-induced allergic airway inflammation without a significant effect on the Th2/Th1/Th17 balance in wild-type mice. In conclusion, the overexpression of miR-146a has a strong anti-inflammatory effect on RV infection in HBECs and a mouse model of allergic airway inflammation, while a lack of miR-146a/b leads to attenuated type 2 cell responses in mouse models of allergic airway inflammation and RV-induced exacerbation of allergic airway inflammation. Furthermore, our data indicate that the application of CPP-miR-146a nanocomplexes has therapeutic potential for targeting airway inflammation.


Subject(s)
Asthma/pathology , Hypersensitivity/pathology , Inflammation/pathology , MicroRNAs/genetics , Picornaviridae Infections/complications , Th2 Cells/immunology , Adult , Allergens , Animals , Asthma/etiology , Asthma/metabolism , Disease Models, Animal , Female , Humans , Hypersensitivity/etiology , Hypersensitivity/metabolism , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Picornaviridae Infections/virology , Rhinovirus/physiology
3.
Front Immunol ; 12: 635569, 2021.
Article in English | MEDLINE | ID: mdl-33868260

ABSTRACT

While there is convincing evidence on the role of Aire-positive medullary thymic epithelial cells (mTEC) in the induction of central tolerance, the nature and function of post-Aire mTECs and Hassall's corpuscles have remained enigmatic. Here we summarize the existing data on these late stages of mTEC differentiation with special focus on their potential to contribute to central tolerance induction by triggering the unique pro-inflammatory microenvironment in the thymus. In order to complement the existing evidence that has been obtained from mouse models, we performed proteomic analysis on microdissected samples from human thymic medullary areas at different differentiation stages. The analysis confirms that at the post-Aire stages, the mTECs lose their nuclei but maintain machinery required for translation and exocytosis and also upregulate proteins specific to keratinocyte differentiation and cornification. In addition, at the late stages of differentiation, the human mTECs display a distinct pro-inflammatory signature, including upregulation of the potent endogenous TLR4 agonist S100A8/S100A9. Collectively, the study suggests a novel mechanism by which the post-Aire mTECs and Hassall's corpuscles contribute to the thymic microenvironment with potential cues on the induction of central tolerance.


Subject(s)
Cell Differentiation , Cellular Microenvironment , Central Tolerance , Epithelial Cells/metabolism , Inflammation Mediators/metabolism , Thymus Gland/metabolism , Transcription Factors/metabolism , Animals , Calgranulin A/metabolism , Calgranulin B/metabolism , Child, Preschool , Epithelial Cells/immunology , Humans , Infant , Mice , Proteome , Proteomics , Thymus Gland/immunology , Toll-Like Receptor 4/metabolism , AIRE Protein
4.
J Exp Med ; 216(5): 1027-1037, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30918005

ABSTRACT

The autoimmune regulator (Aire) serves an essential function for T cell tolerance by promoting the "promiscuous" expression of tissue antigens in thymic epithelial cells. Aire is also detected in rare cells in peripheral lymphoid organs, but the identity of these cells is poorly understood. Here, we report that Aire protein-expressing cells in lymph nodes exhibit typical group 3 innate lymphoid cell (ILC3) characteristics such as lymphoid morphology, absence of "classical" hematopoietic lineage markers, and dependence on RORγt. Aire+ cells are more frequent among lineage-negative RORγt+ cells of peripheral lymph nodes as compared with mucosa-draining lymph nodes, display a unique Aire-dependent transcriptional signature, express high surface levels of MHCII and costimulatory molecules, and efficiently present an endogenously expressed model antigen to CD4+ T cells. These findings define a novel type of ILC3-like cells with potent APC features, suggesting that these cells serve a function in the control of T cell responses.


Subject(s)
Antigen-Presenting Cells/immunology , Lymph Nodes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , CD11 Antigens/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Gene Expression Regulation , Histocompatibility Antigens Class II/metabolism , Immunity, Innate , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Transcription, Genetic , AIRE Protein
5.
Ann Anat ; 216: 125-134, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29289711

ABSTRACT

It is well known that bacterial lipopolysaccharide (LPS) induces migration of several cellular populations within the spleen. However, there are no data about the impact of LPS on B and T lymphocytes present in the red pulp. Therefore, we used an experimental model in which we tested the effects of intravenously injected LPS on the molecular, cellular and structural changes of the spleen, with special reference to the red pulp lymphocytes. We discovered that LPS induced a massive relocation of B and T lymphocytes from the splenic red pulp, which was independent of the tumor necrosis factor receptor-1 signaling axis. Early after LPS treatment, quantitative real-time PCR analysis revealed the elevated levels of mRNA encoding numerous chemokines and proinflammatory cytokines (XCL1, CXCL9, CXCL10, CCL3, CCL4, CCL5, CCL17, CCL20, CCL22, TNFα and LTα) which affect the navigation and activities of B and T lymphocytes in the lymphoid tissues. An extreme increase in mRNA levels for CCL20 was detected in the white pulp of the LPS-treated mice. The CCL20-expressing cells were localized in the PALS. Some smaller CCL20-expressing cells were evenly dispersed in the B cell zone. Thus, our study provides new knowledge of how microbial products could be involved in shaping the structure of lymphatic organs.


Subject(s)
Lipopolysaccharides/pharmacology , Lymphocytes/drug effects , Receptors, Tumor Necrosis Factor, Type I/drug effects , Spleen/cytology , Animals , B-Lymphocytes/drug effects , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Chemokines/biosynthesis , Cytokines/biosynthesis , Immunohistochemistry , Lymphocyte Count , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Spleen/drug effects , T-Lymphocytes/drug effects
6.
Eur J Immunol ; 48(3): 464-470, 2018 03.
Article in English | MEDLINE | ID: mdl-29150834

ABSTRACT

Protection against mucocutaneous candidiasis depends on the T helper (Th)17 pathway, as gene defects affecting its integrity result in inability to clear Candida albicans infection on body surfaces. Moreover, autoantibodies neutralizing Th17 cytokines have been related to chronic candidiasis in a rare inherited disorder called autoimmune polyendocriopathy candidiasis ectodermal dystrophy (APECED) caused by mutations in autoimmune regulator (AIRE) gene. However, the direct pathogenicity of these autoantibodies has not yet been addressed. Here we show that the level of anti-IL17A autoantibodies that develop in aged Aire-deficient mice is not sufficient for conferring susceptibility to oropharyngeal candidiasis. However, patient-derived monoclonal antibodies that cross-react with murine IL-22 increase the fungal burden on C. albicans infected mucosa. Nevertheless, the lack of macroscopically evident infectious pathology on the oral mucosa of infected mice suggests that additional susceptibility factors are needed to precipitate a clinical disease.


Subject(s)
Antibodies, Neutralizing/immunology , Autoantibodies/immunology , Candidiasis, Oral/immunology , Candidiasis, Oral/microbiology , Interleukins/immunology , Animals , Candida albicans/immunology , Candidiasis, Chronic Mucocutaneous/immunology , Candidiasis, Chronic Mucocutaneous/microbiology , Colony Count, Microbial , Cross Reactions , Disease Models, Animal , Disease Susceptibility , Female , Humans , Interleukin-17/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Polyendocrinopathies, Autoimmune/immunology , Th17 Cells/immunology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/immunology , AIRE Protein , Interleukin-22
7.
J Immunol ; 198(5): 1952-1960, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28108558

ABSTRACT

The thymus is a primary lymphoid organ required for the induction and maintenance of central tolerance. The main function of the thymus is to generate an immunocompetent set of T cells not reactive to self. During negative selection in the thymus, thymocytes with autoreactive potential are either deleted or differentiated into regulatory T cells (Tregs). The molecular basis by which the thymus allows high-efficiency Treg induction remains largely unknown. In this study, we report that IFN regulatory factor 4 (Irf4) is highly expressed in murine thymic epithelium and is required to prime thymic epithelial cells (TEC) for effective Treg induction. TEC-specific Irf4 deficiency resulted in a significantly reduced thymic Treg compartment and increased susceptibility to mononuclear infiltrations in the salivary gland. We propose that Irf4 is imperative for thymic Treg homeostasis because it regulates TEC-specific expression of several chemokines and costimulatory molecules indicated in thymocyte development and Treg induction.


Subject(s)
Epithelial Cells/immunology , Homeostasis , Interferon Regulatory Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Animals , Cell Differentiation , Chemokines/genetics , Chemokines/immunology , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Mice , Self Tolerance , Signal Transduction , T-Lymphocytes, Regulatory/physiology , Thymocytes/immunology , Thymus Gland/immunology
8.
Eur J Immunol ; 46(8): 2008-17, 2016 08.
Article in English | MEDLINE | ID: mdl-27214527

ABSTRACT

During normal pregnancy, the thymus undergoes a severe reduction in size and thymocyte output, which may contribute to maternal-fetal tolerance. It is presently unknown whether the pregnancy-induced thymic involution also affects nonlymphoid thymic cell populations and whether these changes in stromal cells play a role in the reduction in thymocyte numbers. Here, we characterize the changes in thymic lymphoid and nonlymphoid cells and show that pregnancy results in a reduction of all major thymic lymphoid cell populations, including the early T-lymphoid progenitors (TLPs) and thymic regulatory T cells. In addition to the thymocytes, the thymic involution also includes all major nonlymphoid cell populations, which show a profound reduction in cell numbers. We also show that during pregnancy, the thymic nonlymphoid cells exhibit decreased expression of chemokines that are essential for TLP homing: CCL25, CXCL12, CCL21, and CCL19. In addition, the expression of these chemokines was substantially downregulated by short-term treatment with progesterone but not estrogen. Collectively, these findings suggest a novel mechanism for the pregnancy-induced reduction in TLP homing and the resulting thymic involution.


Subject(s)
Chemokines, CC/metabolism , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Chemokines, CC/genetics , Estrogens/administration & dosage , Female , Gene Expression , Mice , Mice, Inbred C57BL , Pregnancy , Progesterone/administration & dosage
9.
Mol Immunol ; 72: 92-102, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27011037

ABSTRACT

The differentiation and proper function of thymic epithelial cells (TECs) depend on various tumor necrosis factor superfamily (TNFSF) signals that are needed to maintain the thymic stromal microenvironment. Nevertheless, the direct transcriptional effects of these signals on TECs remain unclear. To address this issue, we stimulated murine embryonic thymus tissue with selected TNFSF ligands and performed a gene expression profiling study. We show that Aire expression is a direct and specific effect of RANKL stimulation, whereas LTß and TNFα are major inducers of chemokines in the thymic stroma and we propose differential NF-κB binding as one possible cause of these gene expression patterns. Our work provides further insight into the complex molecular pathways that shape the thymic microenvironment and maintain central tolerance.


Subject(s)
Cellular Microenvironment , Stromal Cells/cytology , Thymus Gland/cytology , Tumor Necrosis Factors/physiology , Animals , Gene Expression Profiling , Lymphotoxin beta Receptor/biosynthesis , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , NF-kappa B p50 Subunit/biosynthesis , NF-kappa B p50 Subunit/genetics , Organ Culture Techniques , RANK Ligand/genetics , Signal Transduction , Thymus Gland/embryology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factors/genetics
10.
Eur J Immunol ; 45(12): 3246-56, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26364592

ABSTRACT

Autoimmune regulator (Aire) has a unique expression pattern in thymic medullary epithelial cells (mTECs), in which it plays a critical role in the activation of tissue-specific antigens. The expression of Aire in mTECs is activated by receptor activator of nuclear factor κB (RANK) signaling; however, the molecular mechanism behind this activation is unknown. Here, we characterize a conserved noncoding sequence 1 (CNS1) containing two NF-κB binding sites upstream of the Aire coding region. We show that CNS1-deficient mice lack thymic expression of Aire and share several features of Aire-knockout mice, including downregulation of Aire-dependent genes, impaired terminal differentiation of the mTEC population, and reduced production of thymic Treg cells. In addition, we show that CNS1 is indispensable for RANK-induced Aire expression and that CNS1 is activated by NF-κB pathway complexes containing RelA. Together, our results indicate that CNS1 is a critical link between RANK signaling, NF-κB activation, and thymic expression of Aire.


Subject(s)
NF-kappa B/physiology , Thymus Gland/metabolism , Transcription Factors/physiology , Animals , Binding Sites , Epithelial Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor Activator of Nuclear Factor-kappa B/physiology , Signal Transduction , Thymus Gland/cytology , Transcription Factors/genetics , AIRE Protein
11.
Front Immunol ; 5: 51, 2014.
Article in English | MEDLINE | ID: mdl-24592265

ABSTRACT

Studies on autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED) and its mouse model - both caused by mutant AIRE - have greatly advanced the understanding of thymic processes that generate a self-tolerant T-cell repertoire. Much is now known about the molecular mechanisms by which AIRE induces tissue-specific antigen expression in thymic epithelium, and how this leads to negative selection of auto-reactive thymocytes. However, we still do not understand the processes that lead to the activation of any infrequent naïve auto-reactive T-cells exported by AIRE-deficient thymi. Also, the striking phenotypic differences between APECED and its mouse models have puzzled researchers for years. The aim of this review is to suggest explanations for some of these unanswered questions, based on a fresh view of published experiments. We review evidence that auto-reactive T-cells can be activated by the prolonged neonatal lymphopenia that naturally develops in young Aire-deficient mice due to delayed export of mature thymocytes. Lymphopenia-induced proliferation (LIP) helps to fill the empty space; by favoring auto-reactive T-cells, it also leads to lymphocyte infiltration in the same tissues as in day 3 thymectomized animals. The LIP becomes uncontrolled when loss of Aire is combined with defects in genes responsible for anergy induction and Treg responsiveness, or in signaling from the T-cell receptor and homeostatic cytokines. In APECED patients, LIP is much less likely to be involved in activation of naïve auto-reactive T-cells, as humans are born with a more mature immune system than in neonatal mice. We suggest that human AIRE-deficiency presents with different phenotypes because of additional precipitating factors that compound the defective negative selection of potentially autoaggressive tissue-specific thymocytes.

12.
Front Immunol ; 4: 326, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24130560

ABSTRACT

Although the role that Autoimmune Regulator (Aire) plays in the induction of central tolerance is well known, the precise cellular and molecular mechanisms are still unclear and debated. In the prevailing view, Aire serves mainly as a direct inducer of tissue-specific antigens. However, there is a growing amount of evidence suggesting that Aire modulates the differentiation program of medullary thymic epithelial cells, which may directly contribute to the negative selection of self-reactive thymocytes. In addition, Aire has been shown to regulate the expression of many intrathymic chemokines that are required for the proper localization of thymocytes and dendritic cells, and thus are potentially important for direct and indirect self-antigen presentation in the thymic medulla. Further, recent evidence suggests that the induction of certain antigen-specific regulatory T-cells that translocate to tumors and peripheral tissues can be Aire dependent and may contribute to tissue-specific tolerance. This review summarizes the current understanding of the effects of Aire on these alternative mechanisms for the induction of Aire-induced central tolerance.

13.
Eur J Immunol ; 43(3): 580-3, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23404610

ABSTRACT

In the thymus, in order to become MHC-restricted self-tolerant T cells, developing thymocytes need to interact with cortical and medullary thymic epithelial cells (TECs). Although the presence of a common bipotent progenitor for these functionally and structurally distinct epithelial subsets has been clearly established, the initial developmental stages of these bipotent cells have not been well characterized. In this issue of the European Journal of Immunology, Baik et al. [Eur. J. Immunol. 2013.43: 589-594] focus on the phenotypical changes of the early bipotent populations and show how the cortical and medullary markers are sequentially acquired during TEC development. These findings argue against a binary model in which both cortical and medullary lineages diverge simultaneously from lineage-negative TEC progenitors and highlight an unexpected overlap in the phenotypic properties of these bipotent TECs with their lineage-restricted counterparts.


Subject(s)
Antigens, CD/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Transcription Factors/metabolism , Animals , Minor Histocompatibility Antigens , AIRE Protein
14.
Biochem Biophys Res Commun ; 423(1): 32-7, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22613203

ABSTRACT

AIRE (Autoimmune Regulator) has a central role in the transcriptional regulation of self-antigens in medullary thymic epithelial cells, which is necessary for negative selection of autoreactive T cells. Recent data have shown that AIRE can also induce apoptosis, which may be linked to cross-presentation of these self-antigens. Here we studied AIRE-induced apoptosis using AIRE over-expression in a thymic epithelial cell line as well as doxycycline-inducible HEK293 cells. We show that the HSR/CARD domain in AIRE together with a nuclear localization signal is sufficient to induce apoptosis. In the nuclei of AIRE-positive cells, we also found an increased accumulation of a glycolytic enzyme, glyceraldehyde-3-phosphate (GAPDH) reflecting cellular stress and apoptosis. Additionally, AIRE-induced apoptosis was inhibited with an anti-apoptotic agent deprenyl that blocks GAPDH nitrosylation and nuclear translocation. We propose that the AIRE-induced apoptosis pathway is associated with GAPDH nuclear translocation and induction of NO-induced cellular stress in AIRE-expressing cells.


Subject(s)
Apoptosis/physiology , Cell Nucleus/enzymology , DNA Damage , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Oxidative Stress , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Apoptosis/drug effects , Cytoplasm/metabolism , Doxorubicin/pharmacology , Etoposide/pharmacology , HEK293 Cells , Humans , Nitric Oxide/metabolism , Selegiline/pharmacology , Transcription Factors/genetics , AIRE Protein
15.
Front Immunol ; 3(March): 19, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22448160

ABSTRACT

The autoimmune regulator (Aire)-directed ectopic expression of tissue-specific antigens (TSAs) by mature medullary thymic epithelial cells (mTECs) has been viewed as an essential mechanism in the induction of central tolerance. Recent data suggest that the survival of mTECs extends beyond the Aire+ cell population to form the post-Aire mTEC population and Hassall's corpuscles (HCs). The nature and function of these post-Aire epithelial cells and structures, however, have remained unidentified. In this study, we characterized in detail the end-stage development of mTECs and HCs in both Aire-sufficient and Airedeficient mice. In addition, using a transgenic mouse model in which the LacZ reporter gene is under the control of the endogenous Aire promoter, we purified and analyzed the post-Aire mTECs to characterize their function. We showed that the end-stage maturation of mTECs closely resembles that of keratinocytes and that the lack of Aire results in a marked block of mTEC differentiation, which is partially overcome by ligands for RANK and CD40. We also provide evidence that, during mTEC development, Aire is expressed only once and during a limited 1-2 day period. The following loss of Aire expression is accompanied by a quick downregulation of MHC class II and CD80, and of most of the Aire-dependent and Aire-independent TSAs, with the exception of keratinocyte-specific genes. In the final stage of maturation, the mTECs lose their nuclei to become HCs and specifically express desmogleins (DGs) 1 and 3, which, via cross-presentation by APCs, may contribute to tolerance against these pemphigus vulgaris-related TSAs.

16.
Histochem Cell Biol ; 135(6): 593-601, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21611855

ABSTRACT

We have already shown that metallophilic macrophages, which represent an important component in the thymus physiology, are lacking in lymphotoxin-ß receptor-deficient mice. However, further molecular requirements for the development and correct tissue positioning of these cells are unknown. To this end, we studied a panel of mice deficient in different chemokine ligand or receptor genes. In contrast to normal mice, which have these cells localized in the thymic cortico-medullary zone (CMZ) as a distinct row positioned between the cortex and medulla, in plt/plt (paucity of lymph node T cells) mice lacking the functional CCL19/CCL21 chemokines, metallophilic macrophages are not present in the thymic tissue. Interestingly, in contrast to the CCL19/21-deficient thymus, metallophilic macrophages are present in the CCR7-deficient thymus. However, these cells are not appropriately located in the CMZ, but are mostly crowded in central parts of thymic medulla. The double staining revealed that these metallophilic macrophages are CCR7-negative and CXCR3-positive. In the CXCL13-deficient thymus the number, morphology and localization of metallophilic macrophages are normal. Thus, our study shows that CCL19/21 and its possible signaling through CXCR3 are required for the development of thymic metallophilic macrophages, whereas the CXCL13-CXCR5 signaling is not necessary.


Subject(s)
Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Macrophages/metabolism , Receptors, CXCR3/metabolism , Thymus Gland/metabolism , Animals , Macrophages/cytology , Mice , Mice, Knockout , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, CXCR3/genetics , Signal Transduction , Thymus Gland/cytology
17.
Immunol Cell Biol ; 88(1): 50-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19721455

ABSTRACT

The significance of the autoimmune regulator (Aire) transcription regulator in establishing central tolerance has recently been elucidated in great detail. Still, the role of Aire in medullary thymic epithelial cell (mTEC) physiology is not fully understood. To shed more light on this issue, we studied the ultrastructure of mTECs in Aire-deficient thymus. We show that all types of mTECs show ultrastructural signs of activation and increased intracellular traffic, which suggests that in the absence of Aire their physiology is impaired. Type 6 'large' mTECs are fully developed in Aire-deficient mice and more frequent than in the normal thymus. The frequency of type 5 'undifferentiated' mTECs is also increased. Collectively, our results suggest that the role of Aire in the physiology of mTECs could be more profound and not restricted only to the presentation of self-tissue-restricted antigens and/or apoptosis of end-stage fully mature cell types.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Thymus Gland/metabolism , Thymus Gland/ultrastructure , Transcription Factors/metabolism , Animals , Epithelial Cells/immunology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Protein Transport , Thymus Gland/immunology , Transcription Factors/deficiency , Transcription Factors/immunology , AIRE Protein
18.
J Immunol ; 183(12): 7682-91, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19923453

ABSTRACT

Autoimmune regulator (Aire) has been viewed as a central player in the induction of tolerance. This study examines whether Aire can modulate the production of the thymic chemokines involved in corticomedullary migration and thus play a role in intrathymic thymocyte migration and maturation. Aire deficiency resulted in reduced gene expression and protein levels of the CCR4 and CCR7 ligands in whole thymi of mice, as determined by quantitative PCR analysis and ELISA. The expression of the CCR4 ligands coincided with Aire expression in the CD80(high) medullary thymic epithelial cells, whereas the expression of the CCR7 ligands was detected in other cell populations. Also, the expression pattern of the CCR4 and CCR7 ligands follows that of Aire during postnatal but not during embryonic development. In vitro, overexpression of Aire resulted in an up-regulation of selected CCR4 and CCR7 ligands, which induced selective migration of double-positive and single-positive CD4(+) cells. In vivo, Aire deficiency resulted in a diminished emigration of mature CD4(+) T cells from the thymi of 5-day-old mice. In conclusion, Aire regulates the production of CCR4 and CCR7 ligands in medullary thymic epithelial cells and alters the coordinated maturation and migration of thymocytes. These results suggest a novel mechanism behind the Aire-dependent induction of central tolerance.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Migration Inhibition/immunology , Down-Regulation/immunology , Receptors, CCR4/metabolism , Receptors, CCR7/metabolism , Transcription Factors/deficiency , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cell Migration Inhibition/genetics , Down-Regulation/genetics , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factors/genetics , AIRE Protein
19.
Hum Mol Genet ; 18(24): 4699-710, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19744957

ABSTRACT

The Autoimmune Regulator (AIRE) protein is expressed in thymic medullary epithelial cells, where it promotes the ectopic expression of tissue-restricted antigens needed for efficient negative selection of developing thymocytes. Mutations in AIRE cause APECED syndrome, which is characterized by a breakdown of self-tolerance. The molecular mechanism by which AIRE increases the expression of a variety of different genes remains unknown. Here, we studied AIRE-regulated genes using whole genome expression analysis and chromatin immunoprecipitation. We show that AIRE preferentially activates genes that are tissue-specific and characterized by low levels of initial expression in stably transfected HEK293 cell model and mouse thymic medullary epithelial cells. In addition, the AIRE-regulated genes lack active chromatin marks, such as histone H3 trimethylation (H3K4me3) and acetylation (AcH3), on their promoters. We also show that during activation by AIRE, the target genes acquire histone H3 modifications associated with transcription and RNA polymerase II. In conclusion, our data show that AIRE is able to promote ectopic gene expression from chromatin associated with histone modifications characteristic to inactive genes.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Multigene Family , Transcription Factors/metabolism , Transcriptional Activation , Animals , Cell Line , Chromatin Immunoprecipitation , Genome-Wide Association Study , Humans , Mice , Promoter Regions, Genetic , Protein Processing, Post-Translational , RNA Polymerase II/metabolism , Transcription Factors/genetics , AIRE Protein
20.
Histochem Cell Biol ; 131(5): 643-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19148669

ABSTRACT

Thymic metallophilic macrophages represent a significant component in the thymus physiology. Recently, we showed their presence to be dependent on functional lymphotoxin-beta receptor (LT beta R) signaling pathway. However, it is unknown whether the development of metallophilic macrophages also requires the Autoimmune regulator (Aire) transcription factor, as suggested by some studies for medullary thymic epithelial cells, or perhaps the presence of Aire-expressing thymic epithelial cells themselves. Therefore, we investigated the presence of metallophilic macrophages in Aire-deficient thymus. Our study shows that the metallophilic macrophages are fully developed in the Aire-deficient thymus; their development is not regulated via Aire transcription factor and does not require the presence of Aire-expressing epithelial cells. On the contrary, in alymphoplasia (ALY) mice (deficient in nuclear factor-kappaB-inducing kinase, NIK), which we used as negative control, thymic metallophilic macrophages are completely lacking, similarly as in LT beta R-deficient animals. Together, these results show that the development/maintenance of thymic metallophilic macrophages is executed via LT beta R circumventing the Aire transcription factor. Thus, we shed a new light on the molecular requirements for development of these cells and also show that LT beta R pathway is a common developmental regulator of metallophilic macrophages in different lymphatic organs (i.e., thymus and spleen).


Subject(s)
Lymphotoxin beta Receptor/metabolism , Macrophages/metabolism , Protein Serine-Threonine Kinases/metabolism , Thymus Gland/metabolism , Transcription Factors/metabolism , Animals , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Thymus Gland/cytology , Transcription Factors/genetics , AIRE Protein , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...