Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 37: 497-519, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31026413

RESUMEN

During development innate lymphoid cells and specialized lymphocyte subsets colonize peripheral tissues, where they contribute to organogenesis and later constitute the first line of protection while maintaining tissue homeostasis. A few of these subsets are produced only during embryonic development and remain in the tissues throughout life. They are generated through a unique developmental program initiated in lympho-myeloid-primed progenitors, which lose myeloid and B cell potential. They either differentiate into innate lymphoid cells or migrate to the thymus to give rise to embryonic T cell receptor-invariant T cells. At later developmental stages, adaptive T lymphocytes are derived from lympho-myeloid progenitors that colonize the thymus, while lymphoid progenitors become specialized in the production of B cells. This sequence of events highlights the requirement for stratification in the establishment of immune functions that determine efficient seeding of peripheral tissues by a limited number of cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos/fisiología , Células Progenitoras Linfoides/fisiología , Células T Asesinas Naturales/inmunología , Timo/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Microambiente Celular , Citocinas/metabolismo , Humanos , Inmunidad Innata , Activación de Linfocitos , Comunicación Paracrina , Transcriptoma
2.
Nat Immunol ; 18(10): 1139-1149, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28825702

RESUMEN

The molecular events that initiate lymphoid-lineage specification remain unidentified because the stages of differentiation during which lineage commitment occurs are difficult to characterize. We isolated fetal liver progenitor cells undergoing restriction of their differentiation potential toward the T cell-innate lymphoid cell lineage or the B cell lineage. Transcripts that defined the molecular signatures of these two subsets were sequentially upregulated in lympho-myeloid precursor cells and in common lymphoid progenitor cells, respectively, and this preceded lineage restriction; this indicates that T cell-versus-B cell commitment is not a binary fate 'decision'. The T cell-bias and B cell-bias transcriptional programs were frequently co-expressed in common lymphoid progenitor cells and were segregated in subsets biased toward T cell differentiation or B cell differentiation, after interleukin 7 (IL-7) signaling that controlled the number of progenitor cells engaging in T cell differentiation versus B cell differentiation.


Asunto(s)
Linfocitos B/citología , Linaje de la Célula , Hígado/citología , Linfopoyesis , Linfocitos T/citología , Animales , Linfocitos B/metabolismo , Biomarcadores , Diferenciación Celular/genética , Linaje de la Célula/genética , Análisis por Conglomerados , Feto , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Inmunofenotipificación , Interleucina-7/metabolismo , Hígado/embriología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Linfopoyesis/genética , Ratones , Ratones Transgénicos , Transducción de Señal , Linfocitos T/metabolismo , Transcriptoma
3.
Immunol Rev ; 315(1): 54-70, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36869420

RESUMEN

During embryonic development, several independent generations of hematopoietic cells were identified. They occur in the yolk sac and the intra-embryonic major arteries, in a narrow window of development. They arise sequentially, starting with primitive erythrocytes in the yolk sac blood islands, progressing to less differentiated erythromyeloid progenitors still in the yolk sac, and culminating with multipotent progenitors, some of which will generate the adult hematopoietic stem cell compartment. All these cells contribute to the formation of a layered hematopoietic system that reflects adaptative strategies to the fetal environment and the embryo's needs. It is mostly composed, at these stages, of erythrocytes and tissue-resident macrophages both of yolk sac origin, the latter persisting throughout life. We propose that subsets of lymphocytes of embryonic origin derive from a different intra-embryonic generation of multipotent cells occurring before the emergence of hematopoietic stem cell progenitors. These multipotent cells have a limited lifespan and generate cells that provide basic protection against pathogens before the adaptive immune system is functional, contribute to tissue development and homeostasis, and shape the establishment of a functional thymus. Understanding the properties of these cells will impact the understanding of childhood leukemia and of adult autoimmune pathology and thymic involution.


Asunto(s)
Eritrocitos , Células Madre Hematopoyéticas , Embarazo , Femenino , Humanos , Diferenciación Celular , Hematopoyesis
4.
Nat Immunol ; 15(1): 27-35, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24317038

RESUMEN

The generation of T cells depends on the migration of hematopoietic progenitor cells to the thymus throughout life. The identity of the thymus-settling progenitor cells has been a matter of considerable debate. Here we found that thymopoiesis was initiated by a first wave of T cell lineage-restricted progenitor cells with limited capacity for population expansion but accelerated differentiation into mature T cells. They gave rise to αß and γδ T cells that constituted Vγ3(+) dendritic epithelial T cells. Thymopoiesis was subsequently maintained by less-differentiated progenitor cells that retained the potential to develop into B cells and myeloid cells. In that second wave, which started before birth, progenitor cells had high proliferative capacity but delayed differentiation capacity and no longer gave rise to embryonic γδ T cells. Our work reconciles conflicting hypotheses on the nature of thymus-settling progenitor cells.


Asunto(s)
Diferenciación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Linfocitos T/metabolismo , Timo/metabolismo , Animales , Animales Recién Nacidos , Línea Celular , Linaje de la Célula/genética , Células Cultivadas , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Subunidad alfa del Receptor de Interleucina-7/genética , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Timocitos/citología , Timocitos/metabolismo , Timo/citología , Timo/embriología , Factores de Tiempo , Transcriptoma , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
5.
Immunity ; 47(4): 680-696.e8, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045900

RESUMEN

The classical model of hematopoiesis established in the mouse postulates that lymphoid cells originate from a founder population of common lymphoid progenitors. Here, using a modeling approach in humanized mice, we showed that human lymphoid development stemmed from distinct populations of CD127- and CD127+ early lymphoid progenitors (ELPs). Combining molecular analyses with in vitro and in vivo functional assays, we demonstrated that CD127- and CD127+ ELPs emerged independently from lympho-mono-dendritic progenitors, responded differently to Notch1 signals, underwent divergent modes of lineage restriction, and displayed both common and specific differentiation potentials. Whereas CD127- ELPs comprised precursors of T cells, marginal zone B cells, and natural killer (NK) and innate lymphoid cells (ILCs), CD127+ ELPs supported production of all NK cell, ILC, and B cell populations but lacked T potential. On the basis of these results, we propose a "two-family" model of human lymphoid development that differs from the prevailing model of hematopoiesis.


Asunto(s)
Linfocitos B/metabolismo , Células Asesinas Naturales/metabolismo , Células Progenitoras Linfoides/metabolismo , Linfopoyesis/genética , Linfocitos T/metabolismo , Adolescente , Adulto , Animales , Linfocitos B/citología , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad alfa del Receptor de Interleucina-7/genética , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Células Asesinas Naturales/citología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/trasplante , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , Trasplante de Células Madre , Linfocitos T/citología , Trasplante Heterólogo , Adulto Joven
6.
Nat Immunol ; 12(10): 949-58, 2011 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-21909092

RESUMEN

The transcription factor RORγt is required for the development of several innate lymphoid populations, such as lymphoid tissue-inducer cells (LTi cells) and cells that secrete interleukin 17 (IL-17) or IL-22. The progenitor cells as well as the developmental stages that lead to the emergence of RORγt(+) innate lymphoid cells (ILCs) remain undefined. Here we identify the chemokine receptor CXCR6 as an additional marker of the development of ILCs and show that common lymphoid progenitors lost B cell and T cell potential as they successively acquired expression of the integrin α(4)ß(7) and CXCR6. Whereas fetal RORγt(+) cells matured in the fetal liver environment, adult bone marrow-derived RORγt(+) ILCs matured outside the bone marrow, in a Notch2-dependent manner. Therefore, fetal and adult environments influence the differentiation of RORγt(+) cells differently.


Asunto(s)
Feto/inmunología , Linfocitos/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Receptor Notch2/fisiología , Transducción de Señal , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Inmunidad Innata , Integrinas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores CXCR/fisiología , Receptores CXCR6
7.
Blood ; 137(8): 1024-1036, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33025012

RESUMEN

During embryonic development, multiple waves of hematopoietic progenitors with distinct lineage potential are differentially regulated in time and space. Two different waves of thymic progenitors colonize the fetal thymus where they contribute to thymic organogenesis and homeostasis. The origin, the lineage differentiation potential of the first wave, and their relative contribution in shaping the thymus architecture, remained, however, unclear. Here, we show that the first wave of thymic progenitors comprises a unique population of bipotent T and innatel lymphoid cells (T/ILC), generating a lymphoid tissue inducer cells (LTi's), in addition to invariant Vγ5+ T cells. Transcriptional analysis revealed that innate lymphoid gene signatures and, more precisely, the LTi-associated transcripts were expressed in the first, but not in the second, wave of thymic progenitors. Depletion of early thymic progenitors in a temporally controlled manner showed that the progeny of the first wave is indispensable for the differentiation of autoimmune regulator-expressing medullary thymic epithelial cells (mTECs). We further show that these progenitors are of strict hematopoietic stem cell origin, despite the overlap between lymphopoiesis initiation and the transient expression of lymphoid-associated transcripts in yolk sac (YS) erythromyeloid-restricted precursors. Our work highlights the relevance of the developmental timing on the emergence of different lymphoid subsets, required for the establishment of a functionally diverse immune system.


Asunto(s)
Células Progenitoras Linfoides/citología , Linfocitos T/citología , Timo/citología , Timo/embriología , Animales , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Progenitoras Linfoides/metabolismo , Linfopoyesis , Ratones Endogámicos C57BL , Linfocitos T/metabolismo , Timo/metabolismo , Transcriptoma
8.
Cytometry A ; 101(11): 960-969, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35491762

RESUMEN

The fetal liver (FL) is the main hematopoietic organ during embryonic development. The FL is also the unique anatomical site where hematopoietic stem cells expand before colonizing the bone marrow, where they ensure life-long blood cell production and become mostly resting. The identification of the different cell types that comprise the hematopoietic stroma in the FL is essential to understand the signals required for the expansion and differentiation of the hematopoietic stem cells. We used a panel of monoclonal antibodies to identify FL stromal cells in a 5-laser equipped spectral flow cytometry (FCM) analyzer. The "Autofluorescence Finder" of SONY ID7000 software identified two distinct autofluorescence emission spectra. Using autofluorescence as a fluorescence parameter we could assign the two autofluorescent signals to three distinct cell types and identified surface markers that characterize these populations. We found that one autofluorescent population corresponds to hepatoblast-like cells and cholangiocytes whereas the other expresses mesenchymal transcripts and was identified as stellate cells. Importantly, after birth, autofluorescence becomes the unique identifying property of hepatoblast-like cells because mature cholangiocytes are no longer autofluorescent. These results show that autofluorescence used as a parameter in spectral FCM is a useful tool to identify new cell subsets that are difficult to analyze in conventional FCM.


Asunto(s)
Células Madre Hematopoyéticas , Hígado , Embarazo , Femenino , Humanos , Células de la Médula Ósea , Diferenciación Celular , Médula Ósea , Citometría de Flujo
9.
PLoS Biol ; 17(6): e3000335, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31246945

RESUMEN

The assessment of the regenerative capacity of the heart has been compromised by the lack of surface signatures to characterize cardiomyocytes (CMs). Here, combined multiparametric surface marker analysis with single-cell transcriptional profiling and in vivo transplantation identify the main mouse fetal cardiac populations and their progenitors (PRGs). We found that CMs at different stages of differentiation coexist during development. We identified a population of immature heat stable antigen (HSA)/ cluster of differentiation 24 (CD24)+ CMs that persists throughout life and that, unlike other CM subsets, actively proliferates up to 1 week of age and engrafts cardiac tissue upon transplantation. In the adult heart, a discrete population of HSA/CD24+ CMs appears as mononucleated cells that increase in frequency after infarction. Our work identified cell surface signatures that allow the prospective isolation of CMs at all developmental stages and the detection of a subset of immature CMs throughout life that, although at reduced frequencies, are poised for activation in response to ischemic stimuli. This work opens new perspectives in the understanding and treatment of heart pathologies.


Asunto(s)
Antígeno CD24/metabolismo , Linaje de la Célula/fisiología , Miocitos Cardíacos/metabolismo , Animales , Antígeno CD24/fisiología , Diferenciación Celular , Femenino , Corazón/crecimiento & desarrollo , Corazón/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Regeneración/fisiología , Análisis de la Célula Individual
10.
RNA ; 24(12): 1803-1812, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30242063

RESUMEN

The contribution of basal cellular processes to the regulation of tissue homeostasis has just started to be appreciated. However, our knowledge of the modulation of ribosome biogenesis activity in situ within specific lineages remains very limited. This is largely due to the lack of assays that enable quantitation of ribosome biogenesis in small numbers of cells in vivo. We used a technique, named Flow-FISH, combining cell surface antibody staining and flow cytometry with intracellular ribosomal RNA (rRNA) FISH, to measure the levels of pre-rRNAs of hematopoietic cells in vivo. Here, we show that Flow-FISH reports and quantifies ribosome biogenesis activity in hematopoietic cell populations, thereby providing original data on this fundamental process notably in rare populations such as hematopoietic stem and progenitor cells. We unravel variations in pre-rRNA levels between different hematopoietic progenitor compartments and during erythroid differentiation. In particular, our data indicate that, contrary to what may be anticipated from their quiescent state, hematopoietic stem cells have significant ribosome biogenesis activity. Moreover, variations in pre-rRNA levels do not correlate with proliferation rates, suggesting that cell type-specific mechanisms might regulate ribosome biogenesis in hematopoietic stem cells and progenitors. Our study contributes to a better understanding of the cellular physiology of the hematopoietic system in vivo in unperturbed situations.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Procesamiento Postranscripcional del ARN/genética , ARN Ribosómico/biosíntesis , Ribosomas/genética , Animales , Diferenciación Celular/genética , Ratones , Precursores del ARN/genética , Proteínas Ribosómicas , Ribosomas/metabolismo
11.
Int J Mol Sci ; 20(21)2019 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-31690060

RESUMEN

Innate lymphoid cells (ILC) are important players of early immune defenses in situations like lymphoid organogenesis or in case of immune response to inflammation, infection and cancer. Th1 and Th2 antagonism is crucial for the regulation of immune responses, however mechanisms are still unclear for ILC functions. ILC2 and NK cells were reported to be both involved in allergic airway diseases and were shown to be able to interplay in the regulation of the immune response. CXCR6 is a common chemokine receptor expressed by all ILC, and its deficiency affects ILC2 and ILC1/NK cell numbers and functions in lungs in both steady-state and inflammatory conditions. We determined that the absence of a specific ILC2 KLRG1+ST2- subset in CXCR6-deficient mice is probably dependent on CXCR6 for its recruitment to the lung under inflammation. We show that despite their decreased numbers, lung CXCR6-deficient ILC2 are even more activated cells producing large amount of type 2 cytokines that could drive eosinophilia. This is strongly associated to the decrease of the lung Th1 response in CXCR6-deficient mice.


Asunto(s)
Subgrupos Linfocitarios/inmunología , Neumonía/inmunología , Receptores CXCR6/metabolismo , Animales , Células Cultivadas , Interferón gamma/genética , Interferón gamma/metabolismo , Ratones , Papaína/toxicidad , Neumonía/etiología , Receptores CXCR6/genética
12.
Development ; 139(19): 3521-30, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22899849

RESUMEN

Hematopoietic stem cells (HSCs), which are defined by their capacity to reconstitute adult conventional mice, are first found in the dorsal aorta after 10.5 days post coitus (dpc) and in the fetal liver at 11 dpc. However, lympho-myeloid hematopoietic progenitors are detected in the dorsal aorta from 9 dpc, raising the issue of their role in establishing adult hematopoiesis. Here, we show that these progenitors are endowed with long-term reconstitution capacity, but only engraft natural killer (NK)-deficient Rag2γc(-/-) mice. This novel population, called here immature HSCs, evolves in culture with thrombopoietin and stromal cells, into HSCs, defined by acquisition of CD45 and MHC-1 expression and by the capacity to reconstitute NK-competent mice. This evolution occurs during ontogeny, as early colonization of fetal liver by immature HSCs precedes that of HSCs. Moreover, organ culture experiments show that immature HSCs acquire, in this environment, the features of HSCs.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/fisiología , Hígado/embriología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Feto/metabolismo , Hematopoyesis/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/fisiología , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo
13.
Blood ; 121(10): 1749-59, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23287858

RESUMEN

Transcription factors orchestrate T-lineage differentiation in the thymus. One critical checkpoint involves Notch1 signaling that instructs T-cell commitment at the expense of the B-lineage program. While GATA-3 is required for T-cell specification, its mechanism of action is poorly understood. We show that GATA-3 works in concert with Notch1 to commit thymic progenitors to the T-cell lineage via 2 distinct pathways. First, GATA-3 orchestrates a transcriptional "repertoire" that is required for thymocyte maturation up to and beyond the pro-T-cell stage. Second, GATA-3 critically suppresses a latent B-cell potential in pro­T cells. As such, GATA-3 is essential to sealing in Notch-induced T-cell fate in early thymocyte precursors by promoting T-cell identity through the repression of alternative developmental options.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Factor de Transcripción GATA3/fisiología , Transducción de Señal/inmunología , Linfocitos T/citología , Timo/citología , Animales , Linfocitos B/inmunología , Células Cultivadas , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Noqueados , Receptor Notch1/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/inmunología , Timo/embriología , Timo/inmunología , Factores de Transcripción/metabolismo
14.
Mediators Inflamm ; 2015: 368427, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26494947

RESUMEN

Innate lymphoid cells are present at mucosal sites and represent the first immune barrier against infections, but what contributes to their circulation and homing is still unclear. Using Rag2(-/-) Cxcr6(Gfp/+) reporter mice, we assessed the expression and role of CXCR6 in the circulation of ILC precursors and their progeny. We identify CXCR6 expressing ILC precursors in the bone marrow and characterize their significant increase in CXCR6-deficient mice at steady state, indicating their partial retention in the bone marrow after CXCR6 ablation. Circulation was also impaired during embryonic life as fetal liver from CXCR6-deficient embryos displayed decreased numbers of ILC3 precursors. When injected, fetal CXCR6-deficient ILC3 precursors also fail to home and reconstitute ILC compartments in vivo. We show that adult intestinal ILC subsets have heterogeneous expression pattern of CXCR6, integrin α 4 ß 7, CD62L, CD69, and CD44, with ILC1 and ILC3 being more likely tissue resident lymphocytes. Intestinal ILC subsets were unchanged in percentages and numbers in both mice. We demonstrate that the ILC frequency is maintained due to a significant increase of ILC peripheral proliferation, as well as an increased proliferation of the in situ ILC precursors to compensate their retention in the bone marrow.


Asunto(s)
Regulación de la Expresión Génica , Linfocitos/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Células de la Médula Ósea/citología , Proliferación Celular , Medios de Cultivo , Citometría de Flujo , Inmunidad Innata/inmunología , Intestinos/inmunología , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Transgénicos , Membrana Mucosa , Receptores CXCR6
15.
J Cell Sci ; 125(Pt 7): 1738-49, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22366456

RESUMEN

Skeletal muscle stem cell fate in adult mice is regulated by crucial transcription factors, including the determination genes Myf5 and Myod. The precise role of Myf5 in regulating quiescent muscle stem cells has remained elusive. Here we show that most, but not all, quiescent satellite cells express Myf5 protein, but at varying levels, and that resident Myf5 heterozygous muscle stem cells are more primed for myogenic commitment compared with wild-type satellite cells. Paradoxically however, heterotypic transplantation of Myf5 heterozygous cells into regenerating muscles results in higher self-renewal capacity compared with wild-type stem cells, whereas myofibre regenerative capacity is not altered. By contrast, Pax7 haploinsufficiency does not show major modifications by transcriptome analysis. These observations provide a mechanism linking Myf5 levels to muscle stem cell heterogeneity and fate by exposing two distinct and opposing phenotypes associated with Myf5 haploinsufficiency. These findings have important implications for how stem cell fates can be modulated by crucial transcription factors while generating a pool of responsive heterogeneous cells.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Haploinsuficiencia/genética , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Factor 5 Regulador Miogénico/genética , Animales , Linaje de la Célula , Ratones , Músculo Esquelético/citología , Factor 5 Regulador Miogénico/deficiencia , Factor 5 Regulador Miogénico/metabolismo , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Fenotipo
16.
Development ; 138(14): 3055-66, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21693521

RESUMEN

Vessels are primarily formed from an inner endothelial layer that is secondarily covered by mural cells, namely vascular smooth muscle cells (VSMCs) in arteries and veins and pericytes in capillaries and veinules. We previously showed that, in the mouse embryo, Msx1(lacZ) and Msx2(lacZ) are expressed in mural cells and in a few endothelial cells. To unravel the role of Msx genes in vascular development, we have inactivated the two Msx genes specifically in mural cells by combining the Msx1(lacZ), Msx2(lox) and Sm22α-Cre alleles. Optical projection tomography demonstrated abnormal branching of the cephalic vessels in E11.5 mutant embryos. The carotid and vertebral arteries showed an increase in caliber that was related to reduced vascular smooth muscle coverage. Taking advantage of a newly constructed Msx1(CreERT2) allele, we demonstrated by lineage tracing that the primary defect lies in a population of VSMC precursors. The abnormal phenotype that ensues is a consequence of impaired BMP signaling in the VSMC precursors that leads to downregulation of the metalloprotease 2 (Mmp2) and Mmp9 genes, which are essential for cell migration and integration into the mural layer. Improper coverage by VSMCs secondarily leads to incomplete maturation of the endothelial layer. Our results demonstrate that both Msx1 and Msx2 are required for the recruitment of a population of neural crest-derived VSMCs.


Asunto(s)
Cabeza/irrigación sanguínea , Cabeza/embriología , Proteínas de Homeodominio/metabolismo , Factor de Transcripción MSX1/metabolismo , Músculo Liso Vascular/citología , Cresta Neural/citología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Citometría de Flujo , Silenciador del Gen , Proteínas de Homeodominio/genética , Inmunohistoquímica , Hibridación in Situ , Factor de Transcripción MSX1/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tamoxifeno/administración & dosificación , Tamoxifeno/farmacología , Tomografía/métodos
17.
J Immunol ; 188(4): 1600-8, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22238456

RESUMEN

How T cell progenitors engage into the γδ or αß T cell lineages is a matter of intense debate. In this study, we analyzed the differentiation potential of single thymocytes from wild-type and TCRγδ-transgenic mice at two sequential early developmental stages. Double-negative (DN) 3 progenitors from both wild-type and transgenic mice retain the capacity to engage into both pathways, indicating that full commitment is only completed after this stage. More importantly, DN2 and DN3 progenitors from TCRγδ transgenic mice have strong biases for opposite fates, indicating that developmentally regulated changes, other than the production of a functional TCR, altered their likelihood to become a γδ or an αß T cell. Thus, unlike the differentiation in other hematopoietic lineages, T cell progenitors did not restrict, but rather switch their differentiation potential as they developed.


Asunto(s)
Receptores de Antígenos de Linfocitos T alfa-beta/análisis , Receptores de Antígenos de Linfocitos T gamma-delta/análisis , Linfocitos T/inmunología , Timocitos/citología , Timocitos/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Timocitos/metabolismo , Timo
18.
J Immunol ; 189(8): 3822-30, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22972921

RESUMEN

Lineage commitment is regulated during hematopoiesis, with stepwise loss of differentiation potential ultimately resulting in lineage commitment. In this study we describe a novel population of B/NK bipotent precursors among common lymphoid progenitors in the fetal liver and the bone marrow. The absence of T cell precursor potential, both in vivo and in vitro, is due to low Notch1 expression and secondary to inhibition of E2A activity by members of the inhibitor of DNA binding (Id) protein family. Our results demonstrate a new, Id protein-dependent, molecular mechanism of Notch1 repression, operative in both fetal and adult common lymphoid progenitors, where T cell potential is selectively inhibited without affecting either the B or NK programs. This study identifies Id proteins as negative regulators of T cell specification, before B and NK commitment, and provides important insights into the transcriptional networks orchestrating hematopoiesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Diferenciación Celular/inmunología , Proteínas de Unión al ADN/antagonistas & inhibidores , Regulación hacia Abajo/inmunología , Receptor Notch1/antagonistas & inhibidores , Células Madre/inmunología , Subgrupos de Linfocitos T/inmunología , Tirosina Quinasa 3 Similar a fms/deficiencia , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Regulación hacia Abajo/genética , Proteína 2 Inhibidora de la Diferenciación/deficiencia , Proteína 2 Inhibidora de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/deficiencia , Proteínas Inhibidoras de la Diferenciación/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Familia de Multigenes/genética , Familia de Multigenes/inmunología , Receptor Notch1/biosíntesis , Receptor Notch1/genética , Células Madre/citología , Células Madre/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Tirosina Quinasa 3 Similar a fms/biosíntesis , Tirosina Quinasa 3 Similar a fms/genética
19.
Cell Rep ; 43(1): 113676, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38217855

RESUMEN

Natural killer (NK) cells are the predominant lymphocyte population in the liver. At the onset of non-alcoholic steatohepatitis (NASH), an accumulation of activated NK cells is observed in the liver in parallel with inflammatory monocyte recruitment and an increased systemic inflammation. Using in vivo and in vitro experiments, we unveil a specific stimulation of NK cell-poiesis during NASH by medullary monocytes that trans-present interleukin-15 (IL-15) and secrete osteopontin, a biomarker for patients with NASH. This cellular dialogue leads to increased survival and maturation of NK precursors that are recruited to the liver, where they dampen the inflammatory monocyte infiltration. The increase in the production of both osteopontin and the IL-15/IL-15Rα complex by bone marrow monocytes is induced by endotoxemia. We propose a tripartite gut-liver-bone marrow axis regulating the immune population dynamics and effector functions during liver inflammation.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Animales , Ratones , Monocitos , Osteopontina , Interleucina-15 , Médula Ósea , Inflamación , Células Asesinas Naturales , Ratones Endogámicos C57BL
20.
Front Immunol ; 15: 1285215, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38629063

RESUMEN

The analytical capability of flow cytometry is crucial for differentiating the growing number of cell subsets found in human blood. This is important for accurate immunophenotyping of patients with few cells and a large number of parameters to monitor. Here, we present a 43-parameter panel to analyze peripheral blood mononuclear cells from healthy individuals using 41 fluorescence-labelled monoclonal antibodies, an autofluorescent channel, and a viability dye. We demonstrate minimal population distortions that lead to optimized population identification and reproducible results. We have applied an advanced approach in panel design, in selection of sample acquisition parameters and in data analysis. Appropriate autofluorescence identification and integration in the unmixing matrix, allowed for resolution of unspecific signals and increased dimensionality. Addition of one laser without assigned fluorochrome resulted in decreased fluorescence spill over and improved discrimination of cell subsets. It also increased the staining index when autofluorescence was integrated in the matrix. We conclude that spectral flow cytometry is a highly valuable tool for high-end immunophenotyping, and that fine-tuning of major experimental steps is key for taking advantage of its full capacity.


Asunto(s)
Colorantes Fluorescentes , Leucocitos Mononucleares , Humanos , Anticuerpos Monoclonales , Recuento de Leucocitos , Luz
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA