Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Sci ; 114(9): 3509-3522, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37421165

RESUMEN

CCL21-Ser, a chemokine encoded by the Ccl21a gene, is constitutively expressed in the thymic epithelial cells and stromal cells of secondary lymphoid organs. It regulates immune cell migration and survival through its receptor CCR7. Herein, using CCL21-Ser-expressing melanoma cells and the Ccl21a-deficient mice, we demonstrated the functional role of cancer cell-derived CCL21-Ser in melanoma growth in vivo. The B16-F10 tumor growth was significantly decreased in Ccl21a-deficient mice compared with that in wild-type mice, indicating that host-derived CCL21-Ser contributes to melanoma proliferation in vivo. In Ccl21a-deficient mice, tumor growth of melanoma cells expressing CCL21-Ser was significantly enhanced, suggesting that CCL21-Ser from melanoma cells promotes tumor growth in the absence of host-derived CCL21-Ser. The increase in tumor growth was associated with an increase in the CCR7+ CD62L+ T cell frequency in the tumor tissue but was inversely correlated with Treg frequency, suggesting that naïve T cells primarily promote tumor growth. Adoptive transfer experiments demonstrated that naïve T cells are preferentially recruited from the blood into tumors with melanoma cell-derived CCL21-Ser expression. These results suggest that CCL21-Ser from melanoma cells promotes the infiltration of CCR7+ naïve T cells into the tumor tissues and creates a tumor microenvironment favorable for melanoma growth.


Asunto(s)
Melanoma , Linfocitos T , Ratones , Animales , Receptores CCR7/metabolismo , Quimiocina CCL21/metabolismo , Melanoma/patología , Microambiente Tumoral
2.
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
3.
J Biol Chem ; 295(6): 1658-1672, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31915251

RESUMEN

The proteasome is a protein-degrading molecular complex that is necessary for protein homeostasis and various biological functions, including cell cycle regulation, signal transduction, and immune response. Proteasome activity is finely regulated by a variety of proteasome-interacting molecules. PITHD1 is a recently described molecule that has a domain putatively capable of interacting with the proteasome. However, it is unknown whether PITHD1 can actually bind to proteasomes and what it does in vivo Here we report that PITHD1 is detected specifically in the spermatids in the testis and the cortical thymic epithelium in the thymus. Interestingly, PITHD1 associates with immunoproteasomes in the testis, but not with thymoproteasomes in the thymus. Mice deficient in PITHD1 exhibit severe male infertility accompanied with morphological abnormalities and impaired motility of spermatozoa. Furthermore, PITHD1 deficiency reduces proteasome activity in the testis and alters the amount of proteins that are important for fertilization capability by the sperm. However, the PITHD1-deficient mice demonstrate no detectable defects in the thymus, including T cell development. Collectively, our results identify PITHD1 as a proteasome-interacting protein that plays a nonredundant role in the male reproductive system.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Fertilización , Eliminación de Gen , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Motilidad Espermática , Espermátides/metabolismo , Testículo/metabolismo
4.
Immunity ; 36(3): 427-37, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22425250

RESUMEN

The thymic medulla provides a specialized microenvironment for the negative selection of T cells, with the presence of autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) during the embryonic-neonatal period being both necessary and sufficient to establish long-lasting tolerance. Here we showed that emergence of the first cohorts of Aire(+) mTECs at this key developmental stage, prior to αß T cell repertoire selection, was jointly directed by Rankl(+) lymphoid tissue inducer cells and invariant Vγ5(+) dendritic epidermal T cell (DETC) progenitors that are the first thymocytes to express the products of gene rearrangement. In turn, generation of Aire(+) mTECs then fostered Skint-1-dependent, but Aire-independent, DETC progenitor maturation and the emergence of an invariant DETC repertoire. Hence, our data attributed a functional importance to the temporal development of Vγ5(+) γδ T cells during thymus medulla formation for αß T cell tolerance induction and demonstrated a Rank-mediated reciprocal link between DETC and Aire(+) mTEC maturation.


Asunto(s)
Células Precursoras de Linfocitos T/citología , Células Precursoras de Linfocitos T/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Factores de Transcripción/inmunología , Animales , Diferenciación Celular/inmunología , Microambiente Celular , Células Epiteliales/inmunología , Femenino , Feto/citología , Feto/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Transducción de Señal/inmunología , Timo/citología , Timo/inmunología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteína AIRE
5.
J Immunol ; 203(4): 881-887, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31235550

RESUMEN

The affinity for TCR interactions with self-peptide/MHC complexes (pMHC) in the thymus critically affects immature thymocytes that newly express TCRs. Previous fetal thymus organ culture experiments have indicated that difference in the affinity for thymic TCR/pMHC interactions not only determines thymocyte fate between positive and negative selection, but also affects Ag responsiveness of positively selected thymocytes. In the current study, we examined whether TCR/pMHC affinity during positive selection in the thymus would further affect Ag responsiveness of mature T cells in the periphery. To do so, OVA peptide variants were in vivo administered to TAP1-deficient OT-I/TCR-transgenic mice in which T cell development was otherwise arrested at CD4+CD8+ thymocytes because of the lack of self-pMHC presentation in thymic APCs. We found that a group of peptide variants induced the transient generation of OT-I CD8+ T cells in the thymus and the periphery. We also noticed that the affinity threshold for positive and negative selection detected in adult mice in vivo was higher than that measured in fetal thymus organ culture experiments in vitro. Interestingly, we further found that the affinity for positively selecting peptides proportionally affected TCR responsiveness of peripheral naive CD8+ T cells. These results indicate that in vivo administration of a peptide can promote T cell selection in the thymus and the affinity for TCR/pMHC interaction during positive selection fine-tunes Ag responsiveness of peripheral T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Autotolerancia/inmunología , Animales , Antígenos/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Transgénicos , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Timo/inmunología
6.
J Proteome Res ; 19(12): 4826-4843, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33185454

RESUMEN

The Human Proteome Project (HPP) consortium aims to functionally characterize the dark proteome. On the basis of the relevance of olfaction in early neurodegeneration, we have analyzed the dark proteome using data mining in public resources and omics data sets derived from the human olfactory system. Multiple dark proteins localize at synaptic terminals and may be involved in amyloidopathies such as Alzheimer's disease (AD). We have characterized the dark PITH domain-containing protein 1 (PITHD1) in olfactory metabolism using bioinformatics, proteomics, in vitro and in vivo studies, and neuropathology. PITHD1-/- mice exhibit olfactory bulb (OB) proteome changes related to synaptic transmission, cognition, and memory. OB PITHD1 expression increases with age in wild-type (WT) mice and decreases in Tg2576 AD mice at late stages. The analysis across 6 neurological disorders reveals that olfactory tract (OT) PITHD1 is specifically upregulated in human AD. Stimulation of olfactory neuroepithelial (ON) cells with PITHD1 alters the ON phosphoproteome, modifies the proliferation rate, and induces a pro-inflammatory phenotype. This workflow applied by the Spanish C-HPP and Human Brain Proteome Project (HBPP) teams across the ON-OB-OT axis can be adapted as a guidance to decipher functional features of dark proteins. Data are available via ProteomeXchange with identifiers PXD018784 and PXD021634.


Asunto(s)
Enfermedad de Alzheimer , Proteoma , Animales , Ratones , Bulbo Olfatorio/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteómica , Olfato/genética
7.
Int Immunol ; 31(3): 119-125, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30476234

RESUMEN

An immunocompetent and self-tolerant pool of naive T cells is formed in the thymus through the process of repertoire selection. T cells that are potentially capable of responding to foreign antigens are positively selected in the thymic cortex and are further selected in the thymic medulla to help prevent self-reactivity. The affinity between T-cell antigen receptors expressed by newly generated T cells and self-peptide-major histocompatibility complexes displayed in the thymic microenvironments plays a key role in determining the fate of developing T cells during thymic selection. Recent advances in our knowledge of the biology of thymic epithelial cells have revealed unique machinery that contributes to positive and negative selection in the thymus. In this article, we summarize recent findings on thymic T-cell selection, focusing on the machinery unique to thymic epithelial cells.


Asunto(s)
Linfocitos T/citología , Linfocitos T/inmunología , Timo/inmunología , Animales , Células Epiteliales/inmunología , Humanos
8.
J Immunol ; 200(4): 1382-1388, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29298829

RESUMEN

The molecular and cellular biology of thymic epithelial cells (TECs) often relies on the analysis of TECs isolated in enzymatically digested single-cell suspensions derived from mouse thymus. Many independent studies have reported that the estimated cellularity of total TECs isolated from one adult mouse is on the order of up to 105 However, these numbers appear extremely small given that the cellularity of total thymocytes exceeds 108 and that TECs play multiple roles in thymocyte development and repertoire formation. In the present study, we aimed to measure the numbers of ß5t-expressing cortical TECs and Aire-expressing medullary TECs in postnatal mouse thymus in situ without enzymatic digestion. The numbers of these TECs were manually counted in individual thymic sections and were three-dimensionally summed throughout the entire thymic lobes. The results show that the cellularity of total TECs in one 5-wk-old female mouse exceeds 106, containing ∼9 × 105 ß5t+ cortical TECs and ∼1.1 × 106 Aire+ medullary TECs. These results suggest that the use of conventional enzymatic digestion methods for the isolation of TECs may have resulted in the underestimation of the cellularity, and possibly the biology, of TECs.


Asunto(s)
Células Epiteliales/citología , Timo/citología , Animales , Recuento de Células , Femenino , Ratones , Ratones Endogámicos C57BL
9.
J Immunol ; 201(2): 516-523, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29784760

RESUMEN

During αß T cell development in the thymus, migration of newly selected CD4+ and CD8+ thymocytes into medullary areas enables tolerance mechanisms to purge the newly selected αß TCR repertoire of autoreactive specificities. Thymic dendritic cells (DC) play key roles in this process and consist of three distinct subsets that differ in their developmental origins. Thus, plasmacytoid DC and Sirpα+ conventional DC type 2 are extrathymically derived and enter into the thymus via their respective expression of the chemokine receptors CCR9 and CCR2. In contrast, although Sirpα- conventional DC type 1 (cDC1) are known to arise intrathymically from immature progenitors, the precise nature of such thymus-colonizing progenitors and the mechanisms controlling their thymus entry are unclear. In this article, we report a selective reduction in thymic cDC1 in mice lacking the chemokine receptor CCR7. In addition, we show that the thymus contains a CD11c+MHC class II-Sirpα-Flt3+ cDC progenitor population that expresses CCR7, and that migration of these cells to the thymus is impaired in Ccr7-/- mice. Moreover, thymic cDC1 defects in Ccr7-/- mice are mirrored in plt/plt mice, with further analysis of mice individually lacking the CCR7 ligands CCL21Ser (Ccl21a-/- ) or CCL19 (Ccl19-/-) demonstrating an essential role for CCR7-CCL21Ser during intrathymic cDC1 development. Collectively, our data support a mechanism in which CCR7-CCL21Ser interactions guide the migration of cDC progenitors to the thymus for correct formation of the intrathymic cDC1 pool.


Asunto(s)
Quimiocina CCL21/metabolismo , Células Dendríticas/metabolismo , Receptores CCR7/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Animales , Movimiento Celular/fisiología , Tolerancia Inmunológica/fisiología , Ratones , Ratones Endogámicos C57BL
10.
Immunol Rev ; 271(1): 10-22, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27088904

RESUMEN

The thymic cortex provides a microenvironment for the development and positive selection of immature T cells. Cortical thymic epithelial cells (cTECs), which structurally and functionally support the thymic cortical microenvironment, originate from endodermal epithelial progenitors that arise in the third pharyngeal pouch. Recent studies have revealed that thymic epithelial progenitors pass through a stage where the cells express cTEC-associated molecules prior to lineage separation into cTECs and medullary TECs (mTECs). Here, we review the molecular signatures of cTECs and highlight the development and developmental potential of cTECs.


Asunto(s)
Diferenciación Celular , Células Epiteliales/fisiología , Linfocitos T/fisiología , Timo/anatomía & histología , Animales , Microambiente Celular , Selección Clonal Mediada por Antígenos , Humanos , Tolerancia Inmunológica , Timo/fisiología
11.
Immunogenetics ; 71(3): 217-221, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30324237

RESUMEN

Positive selection of T cells in the thymus is induced by low-affinity TCR recognition of self-peptide-MHC complexes expressed by cortical thymic epithelial cells (cTECs). cTECs express a specialized type of proteasomes, the thymoproteasome, which generates a unique spectrum of MHC class I-associated peptides and plays a critical role in thymic positive selection of CD8+ T cells. However, it remains unclear how the thymoproteasome contributes to the thymic positive selection. More than 30 years ago, the "peptidic self" hypothesis proposed that TCRs recognize MHC-presented peptides only, without interacting with MHC molecules, which turned out to be incorrect. Interestingly, however, by implying that a set of MHC-associated peptides forms immunological self, this hypothesis also predicted that positive selection in the thymus is the primary immune response to "foreign epitope" peptides during T cell development. The thymoproteasome-dependent unique self-peptides may create those foreign epitope peptides displayed in the thymus for positive selection of T cells.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Fragmentos de Péptidos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Fragmentos de Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Linfocitos T/metabolismo , Timo/metabolismo
12.
Eur J Immunol ; 46(4): 846-56, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26694097

RESUMEN

Intrathymic T-cell development is critically dependent on cortical and medullary thymic epithelial cells (TECs). Both epithelial subsets originate during early thymus organogenesis from progenitor cells that express the thymoproteasome subunit ß5t, a typical feature of cortical TECs. Using in vivo lineage fate mapping, we demonstrate in mice that ß5t(+) TEC progenitors give rise to the medullary TEC compartment early in life but significantly limit their contribution once the medulla has completely formed. Lineage-tracing studies at single cell resolution demonstrate for young mice that the postnatal medulla is expanded from individual ß5t(+) cortical progenitors located at the cortico-medullary junction. These results therefore not only define a developmental window during which the expansion of medulla is efficiently enabled by progenitors resident in the thymic cortex, but also reveal the spatio-temporal dynamics that control the growth of the thymic medulla.


Asunto(s)
Células Epiteliales/citología , Complejo de la Endopetidasa Proteasomal/metabolismo , Linfocitos T/citología , Timo/citología , Timo/embriología , Animales , Diferenciación Celular , Linaje de la Célula/inmunología , Proliferación Celular , Doxiciclina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis/fisiología , Células Madre/citología , Linfocitos T/inmunología
13.
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
14.
J Immunol ; 192(2): 630-40, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337745

RESUMEN

A critical process during thymic development of the T cell repertoire is the induction of self-tolerance. Tolerance in developing T cells is highly dependent on medullary thymic epithelial cells (mTEC), and mTEC development in turn requires signals from mature single-positive thymocytes, a bidirectional relationship termed thymus crosstalk. We show that CD28-CD80/86 and CD40-CD40L costimulatory interactions, which mediate negative selection and self-tolerance, upregulate expression of LTα, LTß, and receptor activator for NF-κB in the thymus and are necessary for medullary development. Combined absence of CD28-CD80/86 and CD40-CD40L results in profound deficiency in mTEC development comparable to that observed in the absence of single-positive thymocytes. This requirement for costimulatory signaling is maintained even in a TCR transgenic model of high-affinity TCR-ligand interactions. CD4 thymocytes maturing in the altered thymic epithelial environment of CD40/CD80/86 knockout mice are highly autoreactive in vitro and are lethal in congenic adoptive transfer in vivo, demonstrating a critical role for these costimulatory pathways in self-tolerance as well as thymic epithelial development. These findings demonstrate that cooperativity between CD28-CD80/86 and CD40-CD40L pathways is required for normal medullary epithelium and for maintenance of self-tolerance in thymocyte development.


Asunto(s)
Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Antígenos CD28/inmunología , Antígenos CD40/inmunología , Ligando de CD40/inmunología , Epitelio/inmunología , Autotolerancia/inmunología , Timocitos/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Células Epiteliales/inmunología , Células Asesinas Naturales/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , FN-kappa B/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Regulación hacia Arriba/inmunología
15.
Proc Natl Acad Sci U S A ; 110(24): 9885-90, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23720310

RESUMEN

The thymus provides multiple microenvironments that are essential for the development and repertoire selection of T lymphocytes. The thymic cortex induces the generation and positive selection of T lymphocytes, whereas the thymic medulla establishes self-tolerance among the positively selected T lymphocytes. Cortical thymic epithelial cells (cTECs) and medullary TECs (mTECs) constitute the major stromal cells that structurally form and functionally characterize the cortex and the medulla, respectively. cTECs and mTECs are both derived from the endodermal epithelium of the third pharyngeal pouch. However, the molecular and cellular characteristics of the progenitor cells for the distinct TEC lineages are unclear. Here we report the preparation and characterization of mice that express the recombinase Cre instead of ß5t, a proteasome subunit that is abundant in cTECs and not detected in other cell types, including mTECs. By crossing ß5t-Cre knock-in mice with loxP-dependent GFP reporter mice, we found that ß5t-Cre-mediated recombination occurs specifically in TECs but not in any other cell types in the mouse. Surprisingly, in addition to cTECs, ß5t-Cre-loxP-mediated GFP expression was detected in almost all mTECs. These results indicate that the majority of mTECs, including autoimmune regulator-expressing mTECs, are derived from ß5t-expressing progenitor cells.


Asunto(s)
Células Epiteliales/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Células Madre/metabolismo , Timo/metabolismo , Animales , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Complejo de la Endopetidasa Proteasomal/genética , Linfocitos T/metabolismo , Timo/citología , Timo/embriología , Factores de Tiempo , Factores de Transcripción/metabolismo , Proteína AIRE
16.
Proc Natl Acad Sci U S A ; 110(52): 21107-12, 2013 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-24324158

RESUMEN

Induction of self-tolerance in developing T cells depends on medullary thymic epithelial cells (mTECs), whose development, in turn, requires signals from single-positive (SP) thymocytes. Thus, the absence of SP thymocytes in Tcra(-/-) mice results in a profound deficiency in mTECs. Here, we have probed the mechanism that underlies this requirement for cross-talk with thymocytes in medullary development. Previous studies have implicated nonclassical NF-κB as a pathway important in the development of mTECs, because mice lacking RelB, NIK, or IKKα, critical components of this pathway, have an almost complete absence of mTECs, with resulting autoimmune pathology. We therefore assessed the effect of selective deletion in TEC of TNF receptor-associated factor 3 (TRAF3), an inhibitor of nonclassical NF-κB signaling. Deletion of TRAF3 in thymic epithelial cells allowed RelB-dependent development of normal numbers of AIRE-expressing mTECs in the complete absence of SP thymocytes. Thus, mTEC development can occur in the absence of cross-talk with SP thymocytes, and signals provided by SP T cells are needed to overcome TRAF3-imposed arrest in mTEC development mediated by inhibition of nonclassical NF-κB. We further observed that TRAF3 deletion is also capable of overcoming all requirements for LTßR and CD40, which are otherwise necessary for mTEC development, but is not sufficient to overcome the requirement for RANKL, indicating a role for RANKL that is distinct from the signals provided by SP thymocytes. We conclude that TRAF3 plays a central role in regulation of mTEC development by imposing requirements for SP T cells and costimulation-mediated cross-talk in generation of the medullary compartment.


Asunto(s)
Diferenciación Celular/inmunología , Receptor Cross-Talk/inmunología , Autotolerancia/inmunología , Linfocitos T/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Timocitos/metabolismo , Animales , Antígenos CD40/genética , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 3 Asociado a Receptor de TNF/deficiencia , Timocitos/inmunología
17.
Eur J Immunol ; 44(10): 2872-5, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25216053

RESUMEN

Cortical thymic epithelial cells (cTECs) and medullary thymic epithelial cells (mTECs), which play essential roles in the establishment of a functionally competent and self-tolerant repertoire of T cells, are derived from common thymic epithelial progenitor cells (pTECs). Recent findings indicate that mTECs are derived from cells that express molecules that are abundant in cTECs rather than mTECs, and provide fresh insight into the characteristics of pTECs and their diversification pathways into TEC subpopulations. In this issue of the European Journal of Immunology, Ribeiro et al. [Eur. J. Immunol. 2014. 44: 2918-2924] focus on CCRL1, an atypical chemokine receptor that is highly expressed by cTECs rather than mTECs, and show that CCRL1-expressing embryonic TECs can give rise to mTECs. Interestingly, Ribeiro et al. further report that a fraction of postnatal mTECs express CCRL1 at a low level, suggesting novel complexity in mTECs.


Asunto(s)
Diferenciación Celular/inmunología , Células Epiteliales/citología , Receptores CCR/biosíntesis , Timo/citología , Timo/crecimiento & desarrollo , Animales
18.
Eur J Immunol ; 44(1): 16-22, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24214487

RESUMEN

Thymic epithelial cells (TECs) provide key instructive signals for T-cell differentiation. Thymic cortical (cTECs) and medullary (mTECs) epithelial cells constitute two functionally distinct microenvironments for T-cell development, which derive from a common bipotent TEC progenitor. While seminal studies have partially elucidated events downstream of bipotent TECs in relation to the emergence of mTECs and their progenitors, the control and timing of the emergence of the cTEC lineage, particularly in relation to that of mTEC progenitors, has remained elusive. In this review, we describe distinct models that explain cTEC/mTEC lineage divergence from common bipotent progenitors. In particular, we summarize recent studies in mice providing evidence that mTECs, including the auto-immune regulator(+) subset, derive from progenitors initially endowed with phenotypic properties typically associated with the cTEC lineage. These observations support a novel "serial progression" model of TEC development, in which progenitors serially acquire cTEC lineage markers, prior to their commitment to the mTEC differentiation pathway. Gaining a better understanding of the phenotypic properties of early stages in TEC progenitor development should help in determining the mechanisms regulating cTEC/mTEC lineage development, and in strategies aimed at thymus reconstitution involving TEC therapy.


Asunto(s)
Células Epiteliales/inmunología , Inmunoterapia/métodos , Células Progenitoras Linfoides/inmunología , Modelos Inmunológicos , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Timo/inmunología , Animales , Autoinmunidad , Comunicación Celular/inmunología , Diferenciación Celular , Linaje de la Célula , Humanos , Inmunoterapia/tendencias , Ratones
19.
Curr Top Microbiol Immunol ; 373: 1-17, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23612989

RESUMEN

The thymic cortex provides a microenvironment that supports the generation and T cell antigen receptor (TCR)-mediated selection of CD4(+)CD8(+)TCRαß(+) thymocytes. Cortical thymic epithelial cells (cTECs) are the essential component that forms the architecture of the thymic cortex and induces the generation as well as the selection of newly generated T cells. Here we summarize current knowledge on the development, function, and heterogeneity of cTECs, focusing on the expression and function of ß5t, a cTEC-specific subunit of the thymoproteasome.


Asunto(s)
Células Epiteliales/fisiología , Timo/citología , Animales , Linaje de la Célula , Células Madre Hematopoyéticas/citología , Humanos , Interleucina-7/fisiología , Complejo de la Endopetidasa Proteasomal/análisis , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T/inmunología
20.
J Immunol ; 190(10): 5110-7, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23585674

RESUMEN

Medullary thymic epithelial cells (mTECs) play a pivotal role in the establishment of self-tolerance in T cells by ectopically expressing various tissue-restricted self-Ags and by chemoattracting developing thymocytes. The nuclear protein Aire expressed by mTECs contributes to the promiscuous expression of self-Ags, whereas CCR7-ligand (CCR7L) chemokines expressed by mTECs are responsible for the attraction of positively selected thymocytes. It is known that lymphotoxin signals from the positively selected thymocytes preferentially promote the expression of CCR7L rather than Aire in postnatal mTECs. However, it is unknown how lymphotoxin signals differentially regulate the expression of CCR7L and Aire in mTECs and whether CCR7L-expressing mTECs and Aire-expressing mTECs are distinct populations. In this study, we show that the majority of postnatal mTECs that express CCL21, a CCR7L chemokine, represent an mTEC subpopulation distinct from the Aire-expressing mTEC subpopulation. Interestingly, the development of CCL21-expressing mTECs, but not Aire-expressing mTECs, is impaired in mice deficient in the lymphotoxin ß receptor. These results indicate that postnatal mTECs consist of heterogeneous subsets that differ in the expression of CCL21 and Aire, and that lymphotoxin ß receptor regulates the development of the CCL21-expressing subset rather than the Aire-expressing subset of postnatal mTECs.


Asunto(s)
Quimiocina CCL21/biosíntesis , Receptor beta de Linfotoxina/metabolismo , Timo/metabolismo , Animales , Diferenciación Celular , Células Epiteliales , Receptor beta de Linfotoxina/deficiencia , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR7 , Transducción de Señal , Factores de Transcripción , Proteína AIRE
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA