Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 210(11): 1653-1666, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37067332

RESUMEN

Regulatory T cells (Tregs) are produced in the thymus to establish self-tolerance, and agonistic stimuli by self-Ags play a pivotal role in this process. Although two types of APCs, medullary thymic epithelial cells (mTECs) and dendritic cells (DCs), are responsible for presenting self-Ags together with costimulatory/cytokine signals, the distinct role of each APC in producing Tregs remains enigmatic. We have approached this issue by depleting the mTECs and DCs using mice expressing diphtheria toxin receptors driven by Aire and CD11c promoters, respectively. Depletion of mTECs showed an effect on Treg production quantitatively and qualitatively more profound than that of DCs followed by the development of distinct organ-specific autoimmune lesions in the hosts. Because self-Ags produced by mTECs are transferable to DCs through a process known as Ag transfer, we monitored the process of Ag transfer using mice expressing GFP from TECs. Although GFP expressed from total TECs was effectively transferred to DCs, GFP expressed from cortical TECs was not, suggesting that mTECs are the predominant source of self-Ags. We also found that GFP expressed not only from mature mTECs but also from immature mTECs was transferred to DCs, suggesting that a broad spectrum of molecules were subjected to Ag transfer during mTEC development. Interestingly, the numbers of recirculating non-Tregs producing IL-2, an important source for Treg expansion in the thymus, were reduced only in the mTEC-depleted mice. These results suggested the cooperative but distinct role of mTECs and DCs in the production of Tregs to avoid autoimmunity.


Asunto(s)
Linfocitos T Reguladores , Timo , Ratones , Animales , Ratones Endogámicos C57BL , Células Epiteliales , Células Dendríticas , Diferenciación Celular
2.
J Immunol ; 208(2): 303-320, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34930780

RESUMEN

The deficiency of Aire, a transcriptional regulator whose defect results in the development of autoimmunity, is associated with reduced expression of tissue-restricted self-Ags (TRAs) in medullary thymic epithelial cells (mTECs). Although the mechanisms underlying Aire-dependent expression of TRAs need to be explored, the physical identification of the target(s) of Aire has been hampered by the low and promiscuous expression of TRAs. We have tackled this issue by engineering mice with augmented Aire expression. Integration of the transcriptomic data from Aire-augmented and Aire-deficient mTECs revealed that a large proportion of so-called Aire-dependent genes, including those of TRAs, may not be direct transcriptional targets downstream of Aire. Rather, Aire induces TRA expression indirectly through controlling the heterogeneity of mTECs, as revealed by single-cell analyses. In contrast, Ccl25 emerged as a canonical target of Aire, and we verified this both in vitro and in vivo. Our approach has illuminated the Aire's primary targets while distinguishing them from the secondary targets.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad/inmunología , Quimiocinas CC/metabolismo , Timo/inmunología , Factores de Transcripción/metabolismo , Animales , Autoinmunidad/genética , Quimiocinas CC/genética , Células Epiteliales/inmunología , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Timo/citología , Factores de Transcripción/genética , Transcripción Genética/genética , Proteína AIRE
3.
Adv Exp Med Biol ; 1444: 19-32, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38467970

RESUMEN

One of the difficulties in studying the pathogenesis of autoimmune diseases is that the disease is multifactorial involving sex, age, MHC, environment, and some genetic factors. Because deficiency of Aire, a transcriptional regulator, is an autoimmune disease caused by a single gene abnormality, Aire is an ideal research target for approaching the enigma of autoimmunity, e.g., the mechanisms underlying Aire deficiency can be studied using genetically modified animals. Nevertheless, the exact mechanisms of the breakdown of self-tolerance due to Aire's dysfunction have not yet been fully clarified. This is due, at least in part, to the lack of information on the exact target genes controlled by Aire. State-of-the-art research infrastructures such as single-cell analysis are now in place to elucidate the essential function of Aire. The knowledge gained through the study of Aire-mediated tolerance should help our understanding of the pathogenesis of autoimmune disease in general.


Asunto(s)
Enfermedades Autoinmunes , Poliendocrinopatías Autoinmunes , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Autoinmunidad/genética , Poliendocrinopatías Autoinmunes/genética , Poliendocrinopatías Autoinmunes/metabolismo , Aprendizaje , Timo
4.
Eur J Immunol ; 52(8): 1366-1368, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35446436

RESUMEN

Vγ6+ γδ T cells develop in the thymus at the perinatal stage and are exclusive IL-17A producers among γδ T cells. The loss of MHC class II led to the expansion of IL-17A+ Vγ6+ γδ T cells in the thymus. Thus, MHC class II in the thymus inhibits the generation of IL-17A+ Vγ6+ γδ T cells.


Asunto(s)
Genes MHC Clase II , Interleucina-17 , Receptores de Antígenos de Linfocitos T gamma-delta , Subgrupos de Linfocitos T , Timo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Timo/citología
5.
Int J Mol Sci ; 24(14)2023 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-37511462

RESUMEN

Immune responses in humanized mice are generally inefficient without co-transplantation of human thymus or HLA transgenes. Previously, we generated humanized mice via the intra-bone marrow injection of CD133+ cord blood cells into irradiated adult immunodeficient mice (IBMI-huNSG mice), which could mount functional immune responses against HTLV-1, although the underlying mechanisms were still unknown. Here, we investigated thymocyte development in IBMI-huNSG mice, focusing on the roles of human and mouse MHC restriction. IBMI-huNSG mice had normal developmental profiles but aberrant thymic structures. Surprisingly, the thymic medulla-like regions expanded after immunization due to enhanced thymocyte expansion in association with the increase in HLA-DR+ cells, including CD205+ dendritic cells (DCs). The organ culture of thymus from immunized IBMI-huNSG mice with a neutralizing antibody to HLA-DR showed the HLA-DR-dependent expansion of CD4 single positive thymocytes. Mature peripheral T-cells exhibited alloreactive proliferation when co-cultured with human peripheral blood mononuclear cells. Live imaging of the thymus from immunized IBMI-huNSG mice revealed dynamic adhesive contacts of human-derived thymocytes and DCs accompanied by Rap1 activation. These findings demonstrate that an increase in HLA-DR+ cells by immunization promotes HLA-restricted thymocyte expansion in humanized mice, offering a unique opportunity to generate humanized mice with ease.


Asunto(s)
Leucocitos Mononucleares , Timocitos , Humanos , Ratones , Animales , Células Presentadoras de Antígenos , Timo , Antígenos HLA-DR , Inmunización
6.
Eur J Immunol ; 51(2): 311-318, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32845012

RESUMEN

Autoimmune regulator+ (Aire) medullary thymic epithelial cells (mTECs) play a critical role in tolerance induction. Several studies demonstrated that Aire+ mTECs differentiate further into Post-Aire cells. Yet, the identification of terminal stages of mTEC maturation depends on unique fate-mapping mouse models. Herein, we resolve this limitation by segmenting the mTEChi (MHCIIhi CD80hi ) compartment into mTECA/hi (CD24- Sca1- ), mTECB/hi (CD24+ Sca1- ), and mTECC/hi (CD24+ Sca1+ ). While mTECA/hi included mostly Aire-expressing cells, mTECB/hi contained Aire+ and Aire- cells and mTECC/hi were mainly composed of cells lacking Aire. The differential expression pattern of Aire led us to investigate the precursor-product relationship between these subsets. Strikingly, transcriptomic analysis of mTECA/hi , mTECB/hi , and mTECC/hi sequentially mirrored the specific genetic program of Early-, Late- and Post-Aire mTECs. Corroborating their Post-Aire nature, mTECC/hi downregulated the expression of tissue-restricted antigens, acquired traits of differentiated keratinocytes, and were absent in Aire-deficient mice. Collectively, our findings reveal a new and simple blueprint to survey late stages of mTEC differentiation.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Epiteliales/inmunología , Queratinocitos/inmunología , Timo/inmunología , Factores de Transcripción/genética , Animales , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/inmunología , Proteína AIRE
7.
Immunol Cell Biol ; 100(5): 371-377, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35313042

RESUMEN

Deficiency for AIRE/Aire in both humans and mice results in the development of organ-specific autoimmune disease. We tested whether augmented and/or dysregulated AIRE/Aire expression might be also prone to the breakdown of self-tolerance. To define the effect of augmented Aire expression on the development of autoimmunity, antigen-specific clonal deletion and production of clonotypic regulatory T cells (Tregs) in the thymus were examined using mice expressing two additional copies of Aire in a heterozygous state (3xAire-knockin mice: 3xAire-KI). We found that both clonal deletion of autoreactive T cells and production of clonotypic Tregs in the thymus from 3xAire-KI were impaired in a T-cell receptor-transgenic system. Furthermore, 3xAire-KI females showed higher scores of experimental autoimmune encephalomyelitis induced by myelin oligodendrocyte glycoprotein than wild-type littermates, suggesting that augmented Aire expression exacerbates organ-specific autoimmunity under disease-prone conditions. In humans, we found that one patient with amyopathic dermatomyositis showed CD3- CD19- cells expressing AIRE in the peripheral blood before the treatment but not during the remission phase treated with immunosuppressive drugs. Thus, not only loss of function of AIRE/Aire but also augmented and/or dysregulated expression of AIRE/Aire should be considered for the pathogenesis of organ-specific autoimmunity. We suggest that further analyses should be pursued to establish a novel link between organ-specific autoimmune disease and dysregulated AIRE expression in clinical settings.


Asunto(s)
Autoinmunidad , Encefalomielitis Autoinmune Experimental , Animales , Supresión Clonal , Femenino , Humanos , Tolerancia Inmunológica , Ratones , Glicoproteína Mielina-Oligodendrócito , Timo
8.
Int Immunol ; 32(2): 117-131, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31586207

RESUMEN

Tissue-specific autoimmune diseases are assumed to arise through malfunction of two checkpoints for immune tolerance: defective elimination of autoreactive T cells in the thymus and activation of these T cells by corresponding autoantigens in the periphery. However, evidence for this model and the outcome of such alterations in each or both of the tolerance mechanisms have not been sufficiently investigated. We studied these issues by expressing human AIRE (huAIRE) as a modifier of tolerance function in NOD mice wherein the defects of thymic and peripheral tolerance together cause type I diabetes (T1D). Additive huAIRE expression in the thymic stroma had no major impact on the production of diabetogenic T cells in the thymus. In contrast, huAIRE expression in peripheral antigen-presenting cells (APCs) rendered the mice resistant to T1D, while maintaining other tissue-specific autoimmune responses and antibody production against an exogenous protein antigen, because of the loss of Xcr1+ dendritic cells, an essential component for activating diabetogenic T cells in the periphery. These results contrast with our recent demonstration that huAIRE expression in both the thymic stroma and peripheral APCs resulted in the paradoxical development of muscle-specific autoimmunity. Our results reveal that tissue-specific autoimmunity is differentially controlled by a combination of thymic function and peripheral tolerance, which can be manipulated by expression of huAIRE/Aire in each or both of the tolerance mechanisms.


Asunto(s)
Autoinmunidad/inmunología , Tolerancia Periférica/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Autoantígenos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Humanos , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Linfocitos T/inmunología , Factores de Transcripción/genética , Proteína AIRE
9.
Int Immunol ; 31(3): 127-139, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30534943

RESUMEN

Hassall's corpuscles (HCs) are composed of cornifying, terminally differentiated medullary thymic epithelial cells (mTECs) that are developed under the control of Aire. Here, we demonstrated that HC-mTECs show features of cellular senescence and produce inflammatory cytokines and chemokines including CXCL5, thereby recruiting and activating neutrophils to produce IL-23 in the thymic medulla. We further indicated that thymic plasmacytoid dendritic cells (pDCs) expressing IL-23 receptors constitutively produced Ifna, which plays a role in single positive (SP) cell maturation, in an Il23a-dependent manner. Neutrophil depletion with anti-Ly6G antibody injection resulted in a significant decrease of Ifna expression in the thymic pDCs, suggesting that thymic neutrophil activation underlies the Ifna expression in thymic pDCs in steady state conditions. A New Zealand White mouse strain showing HC hyperplasia exhibited greater numbers and activation of thymic neutrophils and pDCs than B6 mice, whereas Aire-deficient B6 mice with defective HC development and SP thymocyte maturation showed significantly compromised numbers and activation of these cells. These results collectively suggested that HC-mTECs with cell-senescence features initiate a unique cell activation cascade including neutrophils and pDCs leading to the constitutive IFNα expression required for SP T-cell maturation in the thymic medulla.


Asunto(s)
Senescencia Celular , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interferón-alfa/biosíntesis , Neutrófilos/inmunología , Neutrófilos/metabolismo , Timo/inmunología , Animales , Células Cultivadas , Células Dendríticas/citología , Humanos , Interferón-alfa/metabolismo , Ratones , Ratones Endogámicos , Neutrófilos/citología , Timo/citología
10.
Immunity ; 35(1): 69-81, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21683627

RESUMEN

Toll-like receptor-7 (TLR7) and 9, innate immune sensors for microbial RNA or DNA, have been implicated in autoimmunity. Upon activation, TLR7 and 9 are transported from the endoplasmic reticulum (ER) to endolysosomes for nucleic acid sensing by an ER-resident protein, Unc93B1. Little is known, however, about a role for sensor transportation in controlling autoimmunity. TLR9 competes with TLR7 for Unc93B1-dependent trafficking and predominates over TLR7. TLR9 skewing is actively maintained by Unc93B1 and reversed to TLR7 if Unc93B1 loses preferential binding via a D34A mutation. We here demonstrate that mice harboring a D34A mutation showed TLR7-dependent, systemic lethal inflammation. CD4(+) T cells showed marked differentiation toward T helper 1 (Th1) or Th17 cell subsets. B cell depletion abolished T cell differentiation and systemic inflammation. Thus, Unc93B1 controls homeostatic TLR7 activation by balancing TLR9 to TLR7 trafficking.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Células TH1/metabolismo , Células Th17/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Diferenciación Celular , Células Cultivadas , Inflamación , Depleción Linfocítica , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación/genética , Unión Proteica/genética , Transporte de Proteínas , Células TH1/inmunología , Células TH1/patología , Células Th17/inmunología , Células Th17/patología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
11.
J Immunol ; 201(11): 3244-3257, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389776

RESUMEN

Medullary thymic epithelial cells (mTECs), which express a wide range of tissue-restricted Ags (TRAs), contribute to the establishment of self-tolerance by eliminating autoreactive T cells and/or inducing regulatory T cells. Aire controls a diverse set of TRAs within Aire-expressing cells by employing various transcriptional pathways. As Aire has a profound effect on transcriptomes of mTECs, including TRAs not only at the single-cell but also the population level, we suspected that Aire (Aire+ mTECs) might control the cellular composition of the thymic microenvironment. In this study, we confirmed that this is indeed the case by identifying a novel mTEC subset expressing Ly-6 family protein whose production was defective in Aire-deficient thymi. Reaggregated thymic organ culture experiments demonstrated that Aire did not induce the expression of Ly-6C/Ly-6G molecules from mTECs as Aire-dependent TRAs in a cell-intrinsic manner. Instead, Aire+ mTECs functioned in trans to maintain Ly-6C/Ly-6G+ mTECs. Thus, Aire not only controls TRA expression transcriptionally within the cell but also controls the overall composition of mTECs in a cell-extrinsic manner, thereby regulating the transcriptome from mTECs on a global scale.


Asunto(s)
Células Epiteliales/patología , Timo/fisiología , Factores de Transcripción/metabolismo , Animales , Antígenos Ly/metabolismo , Células Cultivadas , Microambiente Celular , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Factores de Transcripción/genética , Activación Transcripcional , Proteína AIRE
12.
J Immunol ; 199(12): 3959-3971, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29101311

RESUMEN

Aire controls the fate of autoreactive thymocytes (i.e., clonal deletion or development into regulatory T cells [Tregs]) through transcriptional control of the expression of tissue-restricted self-antigens (TRAs) from medullary thymic epithelial cells (mTECs) and bone marrow (BM)-derived cells. Although TRAs expressed by mTECs and BM-derived cells are suggested to complement each other to generate a full spectrum of TRAs, little is known about the relative contribution of TRAs from each component for establishment of self-tolerance. Furthermore, the precise role of Aire in specific types of Aire-expressing APCs remains elusive. We have approached these issues by generating two different types of transgenic mouse (Tg) model, which express a prefixed model self-antigen driven by the insulin promoter or the Aire promoter. In the insulin-promoter Tg model, mTECs alone were insufficient for clonal deletion, and BM-derived APCs were required for this action by utilizing Ag transferred from mTECs. In contrast, mTECs alone were able to induce Tregs, although at a much lower efficiency in the absence of BM-derived APCs. Importantly, lack of Aire in mTECs, but not in BM-derived APCs, impaired both clonal deletion and production of Tregs. In the Aire-promoter Tg model, both mTECs and BM-derived APCs could independently induce clonal deletion without Aire, and production of Tregs was impaired by the lack of Aire in mTECs, but not in BM-derived APCs. These results suggest that the fate of autoreactive thymocytes together with the requirement for Aire depend on the cell types that express self-antigens and the types of APCs involved in tolerance induction.


Asunto(s)
Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Autoantígenos/inmunología , Supresión Clonal/inmunología , Linfocitos T Reguladores/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Autoantígenos/biosíntesis , Autoantígenos/genética , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Técnicas de Sustitución del Gen , Genes Sintéticos , Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/biosíntesis , Ovalbúmina/genética , Ovalbúmina/inmunología , Regiones Promotoras Genéticas , Ratas , Organismos Libres de Patógenos Específicos , Timo/citología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transgenes , Proteína AIRE
13.
J Autoimmun ; 86: 75-92, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28931462

RESUMEN

Autoimmunity is prevented by the function of the autoimmune regulator [AIRE (Aire in mice)], which promotes the expression of a wide variety of tissue-restricted antigens (TRAs) from medullary thymic epithelial cells (mTECs) and from a subset of peripheral antigen-presenting cells (APCs). We examined the effect of additive expression of human AIRE (huAIRE) in a model of autoimmune diabetes in NOD mice. Unexpectedly, we observed that mice expressing augmented AIRE/Aire developed muscle-specific autoimmunity associated with incomplete maturation of mTECs together with impaired expression of Aire-dependent TRAs. This led to failure of deletion of autoreactive T cells together with dramatically reduced production of regulatory T cells in the thymus. In peripheral APCs, expression of costimulatory molecules was augmented. We suggest that levels of Aire expression need to be tightly controlled for maintenance of immunological tolerance. Our results also highlight the importance of coordinated action between central tolerance and peripheral tolerance under the common control of Aire.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Músculos/inmunología , Polimiositis/inmunología , Timo/inmunología , Factores de Transcripción/metabolismo , Animales , Autoantígenos/metabolismo , Autoinmunidad , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Especificidad de Órganos , Factores de Transcripción/genética , Proteína AIRE
14.
Immunity ; 29(3): 423-37, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18799149

RESUMEN

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.


Asunto(s)
Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Células Epiteliales/citología , Células Epiteliales/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Autotolerancia , Timo/citología , Animales , Autoinmunidad , Antígenos CD40/deficiencia , Diferenciación Celular , Células Epiteliales/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ligando RANK/deficiencia , Ligando RANK/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/metabolismo , Timo/embriología , Timo/inmunología , Timo/fisiología , Quinasa de Factor Nuclear kappa B
15.
Immunity ; 29(3): 438-50, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18799150

RESUMEN

The thymic medulla provides a microenvironment where medullary thymic epithelial cells (mTECs) express autoimmune regulator and diverse tissue-restricted genes, contributing to launching self-tolerance. Positive selection is essential for thymic medulla formation via a previously unknown mechanism. Here we show that the cytokine RANK ligand (RANKL) was produced by positively selected thymocytes and regulated the cellularity of mTEC by interacting with RANK and osteoprotegerin. Forced expression of RANKL restored thymic medulla in mice lacking positive selection, whereas RANKL perturbation impaired medulla formation. These results indicate that RANKL produced by positively selected thymocytes is responsible for fostering thymic medulla formation, thereby establishing central tolerance.


Asunto(s)
Células Epiteliales/inmunología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Linfocitos T/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Timo/inmunología , Factores de Transcripción/metabolismo , Animales , Autoinmunidad , Células Epiteliales/citología , Células Epiteliales/metabolismo , Ratones , Osteoprotegerina/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Autotolerancia , Linfocitos T/citología , Linfocitos T/metabolismo , Timo/citología , Timo/metabolismo , Proteína AIRE
16.
J Immunol ; 195(11): 5149-58, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26503950

RESUMEN

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.


Asunto(s)
Células Epiteliales/metabolismo , Timo/metabolismo , Factores de Transcripción/biosíntesis , Animales , Diferenciación Celular , Toxina Diftérica/farmacología , Células Epiteliales/citología , Expresión Génica , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Timo/citología , Factores de Transcripción/genética , Proteína AIRE
17.
J Immunol ; 195(10): 4641-9, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26453754

RESUMEN

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.


Asunto(s)
Autoinmunidad/genética , Autotolerancia/genética , Timocitos/inmunología , Timo/inmunología , Factores de Transcripción/genética , Animales , Autoinmunidad/inmunología , Diferenciación Celular/inmunología , Cromosomas Artificiales Bacterianos/genética , Células Epiteliales/citología , Células Epiteliales/inmunología , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Autotolerancia/inmunología , Linfocitos T/inmunología , Timocitos/citología , Timo/citología , Factores de Transcripción/metabolismo , Proteína AIRE
18.
Eur J Immunol ; 45(12): 3237-40, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26643138

RESUMEN

Aire has been cloned as the gene responsible for a hereditary type of organ-specific autoimmune disease. Aire controls the expression of a wide array of tissue-restricted Ags by medullary thymic epithelial cells (mTECs), thereby leading to clonal deletion and Treg-cell production, and ultimately to the establishment of self-tolerance. However, relatively little is known about the mechanism responsible for the control of Aire expression itself. In this issue of the European Journal of Immunology, Haljasorg et al. [Eur. J. Immunol. 2015. 45: 3246-3256] have reported the presence of an enhancer element for Aire that binds with NF-κB components downstream of the TNF receptor family member, RANK (receptor activator of NF-κB). The results suggest that RANK has a dual mode of action in Aire expression: one involving the promotion of mTEC differentiation and the other involving activation of the molecular switch for Aire within mature mTECs.


Asunto(s)
Factores de Transcripción/fisiología , Animales , Células Epiteliales/fisiología , Humanos , Ratones , FN-kappa B/fisiología , Receptor Activador del Factor Nuclear kappa-B/fisiología , Transducción de Señal , Timo/citología , Factores de Transcripción/genética , Proteína AIRE
19.
J Immunol ; 193(9): 4356-67, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25261487

RESUMEN

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.


Asunto(s)
Comunicación Celular/inmunología , Células Dendríticas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Autotolerancia/inmunología , Timocitos/inmunología , Animales , Antígeno B7-1/metabolismo , Diferenciación Celular , Células Dendríticas/metabolismo , Células Epiteliales/metabolismo , Expresión Génica , Inmunofenotipificación , Masculino , Ratones , Ratones Transgénicos , Modelos Inmunológicos , Mutación , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Células del Estroma/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Timocitos/metabolismo , Timo/inmunología , Timo/metabolismo , Quinasa de Factor Nuclear kappa B
20.
J Immunol ; 192(6): 2585-92, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24516201

RESUMEN

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.


Asunto(s)
Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Células Epiteliales/inmunología , Factores de Transcripción/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Autoantígenos/inmunología , Autoantígenos/metabolismo , Antígeno B7-1/inmunología , Antígeno B7-1/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Reactividad Cruzada/genética , Reactividad Cruzada/inmunología , Células Epiteliales/metabolismo , Citometría de Flujo , Inmunohistoquímica , Cinética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Timocitos/citología , Timocitos/inmunología , Timocitos/metabolismo , Timo/citología , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína AIRE
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA