Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Immunol ; 195(11): 5149-58, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26503950

ABSTRACT

Aire in medullary thymic epithelial cells (mTECs) plays an important role in the establishment of self-tolerance. Because Aire(+) mTECs appear to be a limited subset, they may constitute a unique lineage(s) among mTECs. An alternative possibility is that all mTECs are committed to express Aire in principle, but Aire expression by individual mTECs is conditional. To investigate this issue, we established a novel Aire reporter strain in which endogenous Aire is replaced by the human AIRE-GFP-Flag tag (Aire/hAGF-knockin) fusion gene. The hAGF reporter protein was produced and retained very efficiently within mTECs as authentic Aire nuclear dot protein. Remarkably, snapshot analysis revealed that mTECs expressing hAGF accounted for >95% of mature mTECs, suggesting that Aire expression does not represent a particular mTEC lineage(s). We confirmed this by generating Aire/diphtheria toxin receptor-knockin mice in which long-term ablation of Aire(+) mTECs by diphtheria toxin treatment resulted in the loss of most mature mTECs beyond the proportion of those apparently expressing Aire. These results suggest that Aire expression is inherent to all mTECs but may occur at particular stage(s) and/or cellular states during their differentiation, thus accounting for the broad impact of Aire on the promiscuous gene expression of mTECs.


Subject(s)
Epithelial Cells/metabolism , Thymus Gland/metabolism , Transcription Factors/biosynthesis , Animals , Cell Differentiation , Diphtheria Toxin/pharmacology , Epithelial Cells/cytology , Gene Expression , Gene Expression Regulation , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Heparin-binding EGF-like Growth Factor/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Thymus Gland/cytology , Transcription Factors/genetics , AIRE Protein
2.
J Immunol ; 195(10): 4641-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26453754

ABSTRACT

Cortical thymic epithelial cells (cTECs) and medullary thymic epithelial cells (mTECs) play essential roles in the positive and negative selection of developing thymocytes, respectively. Aire in mTECs plays an essential role in the latter process through expression of broad arrays of tissue-restricted Ags. To determine whether the location of Aire within the medulla is absolutely essential or whether Aire could also function within the cortex for establishment of self-tolerance, we used bacterial artificial chromosome technology to establish a semiknockin strain of NOD-background (ß5t/Aire-transgenic) mice expressing Aire under control of the promoter of ß5t, a thymoproteasome expressed exclusively in the cortex. Although Aire was expressed in cTECs as typical nuclear dot protein in ß5t/Aire-Tg mice, cTECs expressing Aire ectopically did not confer transcriptional expression of either Aire-dependent or Aire-independent tissue-restricted Ag genes. We then crossed ß5t/Aire-Tg mice with Aire-deficient NOD mice, generating a strain in which Aire expression was confined to cTECs. Despite the presence of Aire(+) cTECs, these mice succumbed to autoimmunity, as did Aire-deficient NOD mice. The thymic microenvironment harboring Aire(+) cTECs, within which many Aire-activated genes were present, also showed no obvious alteration of positive selection, suggesting that Aire's unique property of generating a self-tolerant T cell repertoire is functional only in mTECs.


Subject(s)
Autoimmunity/genetics , Self Tolerance/genetics , Thymocytes/immunology , Thymus Gland/immunology , Transcription Factors/genetics , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Chromosomes, Artificial, Bacterial/genetics , Epithelial Cells/cytology , Epithelial Cells/immunology , Gene Knock-In Techniques , Mice , Mice, Inbred NOD , Mice, Knockout , Self Tolerance/immunology , T-Lymphocytes/immunology , Thymocytes/cytology , Thymus Gland/cytology , Transcription Factors/metabolism , AIRE Protein
3.
J Immunol ; 193(9): 4356-67, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25261487

ABSTRACT

Essential roles of NF-κB-inducing kinase (NIK) for the development of medullary thymic epithelial cells (mTECs) and regulatory T cells have been highlighted by studies using a strain of mouse bearing a natural mutation of the NIK gene (aly mice). However, the exact mechanisms underlying the defect in thymic cross-talk leading to the breakdown of self-tolerance in aly mice remain elusive. In this study, we demonstrated that production of regulatory T cells and the final maturation process of positively selected conventional αß T cells are impaired in aly mice, partly because of a lack of mature mTECs. Of note, numbers of thymic dendritic cells and their expression of costimulatory molecules were also affected in aly mice in a thymic stroma-dependent manner. The results suggest a pivotal role of NIK in the thymic stroma in establishing self-tolerance by orchestrating cross-talk between mTECs and dendritic cells as well as thymocytes. In addition, we showed that negative selection was impaired in aly mice as a result of the stromal defect, which accounts for the development of organ-specific autoimmunity through a lack of normal NIK.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Protein Serine-Threonine Kinases/metabolism , Self Tolerance/immunology , Thymocytes/immunology , Animals , B7-1 Antigen/metabolism , Cell Differentiation , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Gene Expression , Immunophenotyping , Male , Mice , Mice, Transgenic , Models, Immunological , Mutation , Phenotype , Protein Serine-Threonine Kinases/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Stromal Cells/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , NF-kappaB-Inducing Kinase
4.
J Immunol ; 192(6): 2585-92, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24516201

ABSTRACT

Understanding the cellular dynamics of Aire-expressing lineage(s) among medullary thymic epithelial cells (AEL-mTECs) is essential for gaining insight into the roles of Aire in establishment of self-tolerance. In this study, we monitored the maturation program of AEL-mTECs by temporal lineage tracing, in which bacterial artificial chromosome transgenic mice expressing tamoxifen-inducible Cre recombinase under control of the Aire regulatory element were crossed with reporter strains. We estimated that the half-life of AEL-mTECs subsequent to Aire expression was ∼7-8 d, which was much longer than that reported previously, owing to the existence of a post-Aire stage. We found that loss of Aire did not alter the overall lifespan of AEL-mTECs, inconsistent with the previous notion that Aire expression in medullary thymic epithelial cells (mTECs) might result in their apoptosis for efficient cross-presentation of self-antigens expressed by AEL-mTECs. In contrast, Aire was required for the full maturation program of AEL-mTECs, as exemplified by the lack of physiological downregulation of CD80 during the post-Aire stage in Aire-deficient mice, thus accounting for the abnormally increased CD80(high) mTECs seen in such mice. Of interest, increased CD80(high) mTECs in Aire-deficient mice were not mTEC autonomous and were dependent on cross-talk with thymocytes. These results further support the roles of Aire in the differentiation program of AEL-mTECs.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Epithelial Cells/immunology , Transcription Factors/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Autoantigens/immunology , Autoantigens/metabolism , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Cross-Priming/genetics , Cross-Priming/immunology , Epithelial Cells/metabolism , Flow Cytometry , Immunohistochemistry , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/cytology , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , AIRE Protein
5.
J Immunol ; 186(9): 5047-57, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21441458

ABSTRACT

It has recently become clear that signals mediated by members of the TNFR superfamily, including lymphotoxin-ß receptor (LTßR), 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 LTßR signal upregulates expression of RANK in the thymic stroma, thereby promoting accessibility to the RANK ligand necessary for mTEC differentiation. Cooperation between the LTßR 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 LTßR and CD40 signals. Thus, the LTßR 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.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/cytology , Lymphotoxin beta Receptor/metabolism , Organogenesis , Receptor Activator of Nuclear Factor-kappa B/biosynthesis , Signal Transduction , Thymus Gland/embryology , Animals , CD40 Antigens/metabolism , Embryo, Mammalian , Flow Cytometry , Gene Expression , Gene Expression Regulation , Immunohistochemistry , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Self Tolerance/immunology , Thymus Gland/cytology , Thymus Gland/metabolism
6.
Proc Natl Acad Sci U S A ; 105(18): 6720-4, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18436651

ABSTRACT

Controlled proteolytic degradation of specialized junctional structures, corneodesmosomes, by epidermal proteases is an essential process for physiological desquamation of the skin. Corneodesmosin (CDSN) is an extracellular component of corneodesmosomes and, although considerable debate still exists, genetic studies have suggested that the CDSN gene in the major psoriasis-susceptibility locus (PSORS1) may be responsible for susceptibility to psoriasis, a human skin disorder characterized by excessive growth and aberrant differentiation of keratinocytes. CDSN is also expressed in the inner root sheath of hair follicles, and a heterozygous nonsense mutation of the CDSN gene in humans is associated with scalp-specific hair loss of poorly defined etiology. Here, we have investigated the pathogenetic roles of CDSN loss of function in the development of skin diseases by generating a mouse strain with targeted deletion of the Cdsn gene. Cdsn-deficient mouse skin showed detachment of the stratum corneum from the underlying granular layer and/or detachment within the upper granular layers due to the disrupted integrity of the corneodesmosomes. When grafted onto immunodeficient mice, Cdsn-deficient skin showed rapid hair loss together with epidermal abnormalities resembling psoriasis. These results underscore the essential roles of CDSN in hair physiology and suggest functional relevance of CDSN gene polymorphisms to psoriasis susceptibility.


Subject(s)
Gene Deletion , Gene Targeting , Glycoproteins/genetics , Hair/physiology , Skin Physiological Phenomena/genetics , Animals , Desmosomes/metabolism , Desmosomes/pathology , Glycoproteins/deficiency , Hair/growth & development , Mice , Phenotype , Psoriasis/pathology , Skin Abnormalities/genetics , Skin Abnormalities/ultrastructure , Skin Transplantation
7.
J Clin Invest ; 116(5): 1292-301, 2006 May.
Article in English | MEDLINE | ID: mdl-16628255

ABSTRACT

Factors that determine the spectrum of target organs involved in autoimmune destruction are poorly understood. Although loss of function of autoimmune regulator (AIRE) in thymic epithelial cells is responsible for autoimmunity, the pathogenic roles of AIRE in regulating target-organ specificity remain elusive. In order to gain insight into this issue, we have established NOD mice, an animal model of type 1 diabetes caused by autoimmune attack against beta cell islets, in which Aire has been abrogated. Remarkably, acinar cells rather than beta cell islets were the major targets of autoimmune destruction in Aire-deficient NOD mice, and this alteration of intra-pancreatic target-organ specificity was associated with production of autoantibody against pancreas-specific protein disulfide isomerase (PDIp), an antigen expressed predominantly by acinar cells. Consistent with this pathological change, the animals were resistant to the development of diabetes. The results suggest that Aire not only is critical for the control of self-tolerance but is also a strong modifier of target-organ specificity through regulation of T cell repertoire diversification. We also demonstrated that transcriptional expression of PDIp was retained in the Aire-deficient NOD thymus, further supporting the concept that Aire may regulate the survival of autoreactive T cells beyond transcriptional control of self-protein expression in the thymus.


Subject(s)
Pancreas/pathology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Autoimmune Diseases/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/prevention & control , Disease Models, Animal , Female , Genetic Predisposition to Disease , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/metabolism , AIRE Protein
8.
J Exp Med ; 207(5): 963-71, 2010 May 10.
Article in English | MEDLINE | ID: mdl-20404099

ABSTRACT

The roles of autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) in the organization of the thymic microenvironment for establishing self-tolerance are enigmatic. We sought to monitor the production and maintenance of Aire-expressing mTECs by a fate-mapping strategy in which bacterial artificial chromosome transgenic (Tg) mice expressing Cre recombinase under the control of the Aire regulatory element were crossed with a GFP reporter strain. We found that, in addition to its well recognized expression within mature mTECs, Aire was expressed in the early embryo before emergence of the three germ cell layers. This observation may help to explain the development of ectodermal dystrophy often seen in patients with AIRE deficiency. With the use of one Tg line in which Cre recombinase expression was confined to mTECs, we found that Aire(+)CD80(high) mTECs further progressed to an Aire(-)CD80(intermediate) stage, suggesting that Aire expression is not constitutive from after its induction until cell death but instead is down-regulated at the beginning of terminal differentiation. We also demonstrated that many mTECs of Aire-expressing lineage are in close contact with thymic dendritic cells. This close proximity may contribute to transfer of tissue-restricted self-antigens expressed by mTECs to professional antigen-presenting cells.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/cytology , Thymus Gland/cytology , Thymus Gland/physiology , Transcription Factors/genetics , Animals , Embryonic Development/genetics , Epithelial Cells/physiology , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Lymphoid Tissue/physiology , Male , Mice , Mice, Transgenic , Spermatogenesis/physiology , Testis/physiology , AIRE Protein
9.
J Cell Biol ; 186(3): 393-407, 2009 Aug 10.
Article in English | MEDLINE | ID: mdl-19667129

ABSTRACT

Expression of the p16(Ink4a) tumor suppressor gene, a sensor of oncogenic stress, is up-regulated by a variety of potentially oncogenic stimuli in cultured primary cells. However, because p16(Ink4a) expression is also induced by tissue culture stress, physiological mechanisms regulating p16(Ink4a) expression remain unclear. To eliminate any potential problems arising from tissue culture-imposed stress, we used bioluminescence imaging for noninvasive and real-time analysis of p16(Ink4a) expression under various physiological conditions in living mice. In this study, we show that oncogenic insults such as ras activation provoke epigenetic derepression of p16(Ink4a) expression through reduction of DNMT1 (DNA methyl transferase 1) levels as a DNA damage response in vivo. This pathway is accelerated in the absence of p53, indicating that p53 normally holds the p16(Ink4a) response in check. These results unveil a backup tumor suppressor role for p16(Ink4a) in the event of p53 inactivation, expanding our understanding of how p16(Ink4a) expression is regulated in vivo.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
10.
J Immunol ; 179(10): 6799-807, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982070

ABSTRACT

Signal transduction pathways regulating NF-kappaB activation essential for microenvironment formation in secondary lymphoid organs remain to be determined. We investigated the effect of a deficiency of TNFR-associated factor 6 (TRAF6), which activates the classical NF-kappaB pathway, in splenic microenvironment formation. Two-week-old TRAF6-deficient mice showed severe defects in B cell follicle and marginal zone formation, similar to mutant mice defective in lymphotoxin (Lt) beta receptor (LtbetaR) signal induction of nonclassical NF-kappaB activation. However, analysis revealed a TRAF6 role in architecture formation distinct from its role in the early neonatal Lt signaling pathway. LtbetaR signal was essential for primary B cell cluster formation with initial differentiation of follicular dendritic cells (FDCs) in neonatal mice. In contrast, TRAF6 was dispensable for progression to this stage but was required for converting B cell clusters to B cell follicles and maintaining FDCs through to later stages. Fetal liver transfer experiments suggested that TRAF6 in radiation-resistant cells is responsible for follicle formation. Despite FDC-specific surface marker expression, FDCs in neonatal TRAF6-deficient mice had lost the capability to express CXCL13. These data suggest that developmentally regulated activation of TRAF6 in FDCs is required for inducing CXCL13 expression to maintain B cell follicles.


Subject(s)
Chemokine CXCL13/immunology , Dendritic Cells, Follicular/immunology , Lymphotoxin-beta/immunology , Signal Transduction/immunology , Spleen/immunology , TNF Receptor-Associated Factor 6/immunology , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/immunology , Cell Differentiation , Chemokine CXCL13/biosynthesis , Dendritic Cells, Follicular/metabolism , Liver/growth & development , Liver/immunology , Liver/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/biosynthesis , Lymphotoxin-beta/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Mutant Strains , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Signal Transduction/genetics , Spleen/growth & development , Spleen/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism
11.
Proc Natl Acad Sci U S A ; 104(38): 15034-9, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17848507

ABSTRACT

Although the role of p21(Waf1/Cip1) gene expression is well documented in various cell culture studies, its in vivo roles are poorly understood. To gain further insight into the role of p21(Waf1/Cip1) gene expression in vivo, we attempted to visualize the dynamics of p21(Waf1/Cip1) gene expression in living animals. In this study, we established a transgenic mice line (p21-p-luc) expressing the firefly luciferase under the control of the p21(Waf1/Cip1) gene promoter. In conjunction with a noninvasive bioluminescent imaging technique, p21-p-luc mice enabled us to monitor the endogenous p21(Waf1/Cip1) gene expression in vivo. By monitoring and quantifying the p21(Waf1/Cip1) gene expression repeatedly in the same mouse throughout its entire lifespan, we were able to unveil the dynamics of p21(Waf1/Cip1) gene expression in the aging process. We also applied this system to chemically induced skin carcinogenesis and found that the levels of p21(Waf1/Cip1) gene expression rise dramatically in benign skin papillomas, suggesting that p21(Waf1/Cip1) plays a preventative role(s) in skin tumor formation. Surprisingly, moreover, we found that the level of p21(Waf1/Cip1) expression strikingly increased in the hair bulb and oscillated with a 3-week period correlating with hair follicle cycle progression. Notably, this was accompanied by the expression of p63 but not p53. This approach, together with the analysis of p21(Waf1/Cip1) knockout mice, has uncovered a novel role for the p21(Waf1/Cip1) gene in hair development. These data illustrate the unique utility of bioluminescence imaging in advancing our understanding of the timing and, hence, likely roles of specific gene expression in higher eukaryotes.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Luminescent Measurements/methods , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Firefly Luciferin/metabolism , Fluorescent Antibody Technique , Gene Expression , Luciferases/genetics , Luciferases/metabolism , Luminescent Agents/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
12.
J Immunol ; 176(7): 3995-4002, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16547234

ABSTRACT

IkappaB kinase (IKK) alpha exhibits diverse biological activities through protein kinase-dependent and -independent functions, the former mediated predominantly through a noncanonical NF-kappaB activation pathway. The in vivo function of IKKalpha, however, still remains elusive. Because a natural strain of mice with mutant NF-kappaB-inducing kinase (NIK) manifests autoimmunity as a result of disorganized thymic structure with abnormal expression of Rel proteins in the thymic stroma, we speculated that the NIK-IKKalpha axis might constitute an essential step in the thymic organogenesis that is required for the establishment of self-tolerance. An autoimmune disease phenotype was induced in athymic nude mice by grafting embryonic thymus from IKKalpha-deficient mice. The thymic microenvironment that caused autoimmunity in an IKKalpha-dependent manner was associated with defective processing of NF-kappaB2, resulting in the impaired development of thymic epithelial cells. Thus, our results demonstrate a novel function for IKKalpha in thymic organogenesis for the establishment of central tolerance that depends on its protein kinase activity in cooperation with NIK.


Subject(s)
I-kappa B Kinase/metabolism , Organogenesis , Self Tolerance/immunology , Thymus Gland/embryology , Thymus Gland/immunology , Animals , Gene Expression Regulation , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , Mice , Mice, Knockout , Mutation/genetics , NF-kappa B/metabolism , Oncogene Proteins v-rel/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Thymus Gland/enzymology , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL