Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Biol ; 17(3): e2006716, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30856173

RESUMEN

The interdependence of selective cues during development of regulatory T cells (Treg cells) in the thymus and their suppressive function remains incompletely understood. Here, we analyzed this interdependence by taking advantage of highly dynamic changes in expression of microRNA 181 family members miR-181a-1 and miR-181b-1 (miR-181a/b-1) during late T-cell development with very high levels of expression during thymocyte selection, followed by massive down-regulation in the periphery. Loss of miR-181a/b-1 resulted in inefficient de novo generation of Treg cells in the thymus but simultaneously permitted homeostatic expansion in the periphery in the absence of competition. Modulation of T-cell receptor (TCR) signal strength in vivo indicated that miR-181a/b-1 controlled Treg-cell formation via establishing adequate signaling thresholds. Unexpectedly, miR-181a/b-1-deficient Treg cells displayed elevated suppressive capacity in vivo, in line with elevated levels of cytotoxic T-lymphocyte-associated 4 (CTLA-4) protein, but not mRNA, in thymic and peripheral Treg cells. Therefore, we propose that intrathymic miR-181a/b-1 controls development of Treg cells and imposes a developmental legacy on their peripheral function.


Asunto(s)
MicroARNs/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Citometría de Flujo , Ratones , Ratones Noqueados , MicroARNs/genética , Microscopía Confocal , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Timocitos/metabolismo
2.
Eur J Immunol ; 49(1): 121-132, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30281154

RESUMEN

The interdependence of posttranscriptional gene regulation via miRNA and transcriptional regulatory networks in lymphocyte development is poorly understood. Here, we identified miR-191 as direct upstream modulator of a transcriptional module comprising the transcription factors Foxp1, E2A, and Egr1. Deletion as well as ectopic expression of miR-191 resulted in developmental arrest in B lineage cells, indicating that fine tuning of the combined expression levels of Foxp1, E2A, and Egr1, which in turn control somatic recombination and cytokine-driven expansion, constitutes a prerequisite for efficient B-cell development. In conclusion, we propose that miR-191 acts as a rheostat in B-cell development by fine tuning a key transcriptional program.


Asunto(s)
Linfocitos B/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factores de Transcripción Forkhead/metabolismo , MicroARNs/genética , Proteínas Represoras/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Factores de Transcripción Forkhead/genética , Redes Reguladoras de Genes , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Recombinación Genética , Proteínas Represoras/genética , Transcripción Genética , Transgenes/genética
3.
PLoS One ; 13(2): e0192230, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29389970

RESUMEN

The increased susceptibility to infections of neonates is caused by an immaturity of the immune system as a result of both qualitative and quantitative differences between neonatal and adult immune cells. With respect to B cells, neonatal antibody responses are known to be decreased. Accountable for this is an altered composition of the neonatal B cell compartment towards more immature B cells. However, it remains unclear whether the functionality of individual neonatal B cell subsets is altered as well. In the current study we therefore compared phenotypical and functional characteristics of corresponding neonatal and adult B cell subpopulations. No phenotypic differences could be identified with the exception of higher IgM expression in neonatal B cells. Functional analysis revealed differences in proliferation, survival, and B cell receptor signaling. Most importantly, neonatal B cells showed severely impaired class-switch recombination (CSR) to IgG and IgA. This was associated with increased expression of miR-181b in neonatal B cells. Deficiency of miR-181b resulted in increased CSR. With this, our results highlight intrinsic differences that contribute to weaker B cell antibody responses in newborns.


Asunto(s)
Linfocitos B/inmunología , Cambio de Clase de Inmunoglobulina/inmunología , MicroARNs/genética , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Recién Nacido , Ratones , Ratones Endogámicos C57BL
4.
Immunol Cell Biol ; 94(8): 741-6, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27089939

RESUMEN

Expression of microRNA miR-181a/b-1 is critical for intrathymic development of invariant natural killer T (iNKT) cells. However, the underlying mechanism has remained a matter of debate. On the one hand, growing evidence suggested that miR-181a/b-1 is instrumental in setting T-cell receptor (TCR) signaling threshold and thus permits agonist selection of iNKT cells through high-affinity TCR ligands. On the other hand, alterations in metabolic fitness mediated by miR-181a/b-1-dependent dysregulation of phosphatase and tensin homolog (Pten) have been proposed to cause the iNKT-cell defect in miR-181-a/b-1-deficient mice. To re-assess the hypothesis that modulation of TCR signal strength is the key mechanism by which miR-181a/b-1 controls the development of iNKT cells, we generated miR-181a/b-1-deficient mice expressing elevated levels of a Vα14Jα18 TCRα chain. In these mice, development of iNKT cells was fully restored. Furthermore, both subset distribution of iNKT cells as well as TCR Vß repertoire were independent of the presence of miR-181a/b-1 once a Vα14Jα18 TCRα chain was overexpressed. Finally, levels of Pten protein were similar in Vα14Jα18 transgenic mice irrespective of their miR-181a/b-1 status. Collectively, our data support a model in which miR-181 promotes development of iNKT cells primarily by generating a permissive state for agonist selection with alterations in metabolic fitness possibly constituting a secondary effect.


Asunto(s)
MicroARNs/metabolismo , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Animales , Polaridad Celular , Subgrupos Linfocitarios/inmunología , Ratones Transgénicos , MicroARNs/genética , Fosfohidrolasa PTEN/metabolismo , Transgenes
5.
PLoS One ; 10(12): e0145010, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26673421

RESUMEN

Thymic development of αß T lymphocytes into invariant natural killer (NK) T cells depends on their selection via agonistic lipid antigen presented by CD1d. If successful, newly selected NKT cells gain effector functions already in the thymus. Some γδ T cell subsets also acquire effector functions in the thymus. However, it is not clear whether agonistic TCR stimulation is involved in thymic γδ T cell selection and development. Here we combine two genetic models to address this question. MiR-181a/b-1-/-mice, which show impaired agonistic T cell selection of invariant αß NKT cells, were crossed to Tcrd-H2BeGFP reporter mice to monitor selection, intra-thymic expansion and differentiation of γδ T cells. We found that miR-181a/b-1-deficiency had no effect on numbers of thymic γδ T cell or on their differentiation towards an IL-17- or IFN-γ-producing effector phenotype. Also, the composition of peripheral lymph node γδ T cells was not affected by miR-181a/b-1-deficiency. Dendritic epidermal γδ T cells were normally present in knock-out animals. However, we observed elevated frequencies and numbers of γδ NKT cells in the liver, possibly because γδ NKT cells can expand and replace missing αß NKT cells in peripheral niches. In summary, we investigated the role of miR-181a/b-1 for selection, intrathymic development and homeostasis of γδ T cells. We conclude that miR-181a/b-1-dependent modulation of T cell selection is not critically required for innate development of γδ NKT cells or of any other γδ T cell subtypes.


Asunto(s)
Inmunidad Innata , MicroARNs/genética , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Animales , Antígenos CD1d/metabolismo , Diferenciación Celular/genética , Selección Clonal Mediada por Antígenos/genética , Inmunofenotipificación , Hígado/citología , Hígado/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Células T Asesinas Naturales/citología , Fenotipo , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Timo/citología , Timo/inmunología , Timo/metabolismo
6.
J Immunol ; 195(10): 4832-40, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26475928

RESUMEN

miRNAs regulate a large variety of developmental processes including development of the immune system. T cell development is tightly controlled through the interplay of transcriptional programs and cytokine-mediated signals. However, the role of individual miRNAs in this process remains largely elusive. In this study, we demonstrated that hematopoietic cell-specific loss of miR-17∼92, a cluster of six miRNAs implicated in B and T lineage leukemogenesis, resulted in profound defects in T cell development both at the level of prethymic T cell progenitors as well as intrathymically. We identified reduced surface expression of IL-7R and concomitant limited responsiveness to IL-7 signals as a common mechanism resulting in reduced cell survival of common lymphoid progenitors and thymocytes at the double-negative to double-positive transition. In conclusion, we identified miR-17∼92 as a critical modulator of multiple stages of T cell development.


Asunto(s)
Interleucina-7/inmunología , MicroARNs/inmunología , Transducción de Señal/fisiología , Linfocitos T/inmunología , Animales , Animales Modificados Genéticamente , Linfocitos B/citología , Linfocitos B/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Interleucina-7/genética , Ratones , MicroARNs/genética , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Linfocitos T/citología
7.
J Exp Med ; 212(10): 1589-601, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26347471

RESUMEN

Postnatal T cell development depends on continuous colonization of the thymus by BM-derived T lineage progenitors. Both quantitative parameters and the mechanisms of thymus seeding remain poorly understood. Here, we determined the number of dedicated thymus-seeding progenitor niches (TSPNs) capable of supporting productive T cell development, turnover rates of niche occupancy, and feedback mechanisms. To this end, we established multicongenic fate mapping combined with mathematical modeling to quantitate individual events of thymus colonization. We applied this method to study thymus colonization in CCR7(-/-)CCR9(-/-) (DKO) mice, whose TSPNs are largely unoccupied. We showed that ∼160-200 TSPNs are present in the adult thymus and, on average, 10 of these TSPNs were open for recolonization at steady state. Preconditioning of wild-type mice revealed a similar number of TSPNs, indicating that preconditioning can generate space efficiently for transplanted T cell progenitors. To identify potential cellular feedback loops restricting thymus colonization, we performed serial transfer experiments. These experiments indicated that thymus seeding was directly restricted by the duration of niche occupancy rather than long-range effects, thus challenging current paradigms of thymus colonization.


Asunto(s)
Linfocitos T/fisiología , Timo/citología , Animales , Linaje de la Célula , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR/genética , Receptores CCR/metabolismo , Receptores CCR7/genética , Receptores CCR7/metabolismo , Receptores de Interleucina-17/genética , Células Madre/fisiología , Linfocitos T/citología , Timocitos/fisiología , Timo/fisiología , Timo/efectos de la radiación
8.
Blood ; 125(3): 457-64, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25411428

RESUMEN

The origins of dendritic cells (DCs) and other myeloid cells in the thymus have remained controversial. In this study, we assessed developmental relationships between thymic dendritic cells and thymocytes, employing retrovirus-based cellular barcoding and reporter mice, as well as intrathymic transfers coupled with DC depletion. We demonstrated that a subset of early T-lineage progenitors expressed CX3CR1, a bona fide marker for DC progenitors. However, intrathymic transfers into nonmanipulated mice, as well as retroviral barcoding, indicated that thymic dendritic cells and thymocytes were largely of distinct developmental origin. In contrast, intrathymic transfers after in vivo depletion of DCs resulted in intrathymic development of non-T-lineage cells. In conclusion, our data support a model in which the adoption of T-lineage fate by noncommitted progenitors at steady state is enforced by signals from the thymic microenvironment unless niches promoting alternative lineage fates become available.


Asunto(s)
Células Dendríticas/inmunología , Células Mieloides/inmunología , Nicho de Células Madre/inmunología , Células Madre/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Células Dendríticas/citología , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Células Madre/citología , Linfocitos T/citología , Timo/citología
9.
Proc Natl Acad Sci U S A ; 110(18): 7407-12, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589855

RESUMEN

T-cell receptor (TCR) signal strength determines selection and lineage fate at the CD4(+)CD8(+) double-positive stage of intrathymic T-cell development. Members of the miR-181 family constitute the most abundantly expressed microRNA at this stage of T-cell development. Here we show that deletion of miR-181a/b-1 reduced the responsiveness of double-positive thymocytes to TCR signals and virtually abrogated early invariant natural killer T (iNKT) cell development, resulting in a dramatic reduction in iNKT cell numbers in thymus as well as in the periphery. Increased concentrations of agonist ligand rescued iNKT cell development in miR-181a/b-1(-/-) mice. Our results define a critical role of miR-181a/b-1 in early iNKT cell development and show that miR-181a/b-1 sets a TCR signaling threshold for agonist selection.


Asunto(s)
Selección Clonal Mediada por Antígenos/inmunología , MicroARNs/metabolismo , Células T Asesinas Naturales/inmunología , Animales , Proliferación Celular , Ligandos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Células T Asesinas Naturales/citología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
10.
PLoS One ; 7(2): e30864, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355330

RESUMEN

T cell development depends on continuous recruitment of progenitors from bone marrow (BM) to the thymus via peripheral blood. However, both phenotype and functional characteristics of physiological T cell precursors remain ill-defined. Here, we characterized a putative CD135(+)CD27(+) T cell progenitor population, which lacked expression of CD127, CD90, and high levels of CD117 and was therefore termed triple negative precursor (TNP). TNPs were present in both BM and blood and displayed robust T lineage potential, but virtually no myeloid or B lineage potential, in vitro. However, TNPs did not efficiently generate T lineage progeny after intravenous or intrathymic transfer, suggesting that a physiological thymic microenvironment does not optimally support T cell differentiation from TNPs. Thus, we propose that physiological T cell precursors are confined to populations expressing either CD127, CD90, or high levels of CD117 in addition to CD135 and CD27 and that TNPs may have other physiological functions.


Asunto(s)
Médula Ósea/inmunología , Linaje de la Célula , Células Madre Hematopoyéticas/inmunología , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Linfocitos T/inmunología , Antígenos Thy-1/metabolismo , Animales , Médula Ósea/metabolismo , Diferenciación Celular , Células Cultivadas , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Antígenos Comunes de Leucocito/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/citología , Linfocitos T/metabolismo , Timo/citología , Timo/inmunología , Timo/metabolismo
11.
Eur J Immunol ; 41(5): 1256-65, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21425158

RESUMEN

Expression of CX3CR1 is an attribute of myeloid precursors committed to the monocyte/macrophage (Mφ)/DC lineages and is maintained during all stages of DC differentiation. Nevertheless, the exact role of this molecule during developmental progression of myeloid precursors towards the DC lineage remains elusive. To overcome potential compensatory mechanisms and issues of redundancy, we employed competitive adoptive transfer experiments to assess a possible function of CX3CR1 in DC and monocyte/Mφ differentiation in vivo. We show here that expression of CX3CR1 promotes the generation of DCs and monocytes/Mφ under steady-state conditions and during compensatory expansion after selective depletion of DCs, but not under inflammatory conditions evoked by sub-lethal irradiation. Direct administration of CX3CR1-deficient and CX3CR1-sufficient precursors into the spleen or the thymus resulted in a similar competitive advantage of WT over CX3CR1-deficient precursors as i.v. transfer, suggesting that CX3CR1-mediated survival rather than recruitment to lymphoid organs is critical for DC/Mφ differentiation. In conclusion, our data support the hypothesis that CX3CR1 promotes proper development of myeloid precursors into DCs and monocytes/Mφs under steady-state conditions, possibly by providing survival signals or mediating accessibility to organ-specific niches, rather than acting as a mediator of homing to the spleen or the thymus.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/fisiología , Receptores de Quimiocina/metabolismo , Traslado Adoptivo , Animales , Anticuerpos Monoclonales , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular , Células Dendríticas/metabolismo , Células Dendríticas/efectos de la radiación , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inflamación/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Precursoras de Monocitos y Macrófagos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/efectos de la radiación , Transducción de Señal
12.
Blood ; 115(6): 1137-44, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-20009033

RESUMEN

T-cell development in the thymus depends on continuous supply of T-cell progenitors from bone marrow (BM). Several extrathymic candidate progenitors have been described that range from multipotent cells to lymphoid cell committed progenitors and even largely T-lineage committed precursors. However, the nature of precursors seeding the thymus under physiologic conditions has remained largely elusive and it is not known whether there is only one physiologic T-cell precursor population or many. Here, we used a competitive in vivo assay based on depletion rather than enrichment of classes of BM-derived precursor populations, thereby only minimally altering physiologic precursor ratios to assess the contribution of various extrathymic precursors to T-lineage differentiation. We found that under these conditions multiple precursors, belonging to both multipotent progenitor (MPP) and common lymphoid progenitor (CLP) subsets have robust T-lineage potential. However, differentiation kinetics of different precursors varied considerably, which might ensure continuous thymic output despite gated importation of extrathymic precursors. In conclusion, our data suggest that the thymus functions to impose T-cell fate on any precursor capable of filling the limited number of progenitor niches.


Asunto(s)
Diferenciación Celular , Células Precursoras de Linfocitos T/citología , Células Precursoras de Linfocitos T/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Células de la Médula Ósea/inmunología , Linaje de la Célula , Citometría de Flujo , Hematopoyesis , Activación de Linfocitos , Células Progenitoras Linfoides/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Multipotentes/inmunología , Receptores de Interleucina-7/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...