Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
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.
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
4.
Development ; 145(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30042179

RESUMEN

To decipher the populations of cells present in the human fetal pancreas and their lineage relationships, we developed strategies to isolate pancreatic progenitors, endocrine progenitors and endocrine cells. Transcriptome analysis of the individual populations revealed a large degree of conservation among vertebrates in the drivers of gene expression changes that occur at different steps of differentiation, although notably, sometimes, different members of the same gene family are expressed. The transcriptome analysis establishes a resource to identify novel genes and pathways involved in human pancreas development. Single-cell profiling further captured intermediate stages of differentiation and enabled us to decipher the sequence of transcriptional events occurring during human endocrine differentiation. Furthermore, we evaluate how well individual pancreatic cells derived in vitro from human pluripotent stem cells mirror the natural process occurring in human fetuses. This comparison uncovers a few differences at the progenitor steps, a convergence at the steps of endocrine induction, and the current inability to fully resolve endocrine cell subtypes in vitro.


Asunto(s)
Feto/embriología , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/fisiología , Páncreas/embriología , Transcripción Genética/fisiología , Feto/citología , Humanos , Páncreas/citología , Células Madre Pluripotentes/metabolismo
5.
Hum Mol Genet ; 26(3): 599-610, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28025328

RESUMEN

Congenital hypothyroidism is the most common neonatal endocrine disorder and is primarily caused by developmental abnormalities otherwise known as thyroid dysgenesis (TD). We performed whole exome sequencing (WES) in a consanguineous family with TD and subsequently sequenced a cohort of 134 probands with TD to identify genetic factors predisposing to the disease. We identified the novel missense mutations p.S148F, p.R114Q and p.L177W in the BOREALIN gene in TD-affected families. Borealin is a major component of the Chromosomal Passenger Complex (CPC) with well-known functions in mitosis. Further analysis of the missense mutations showed no apparent effects on mitosis. In contrast, expression of the mutants in human thyrocytes resulted in defects in adhesion and migration with corresponding changes in gene expression suggesting others functions for this mitotic protein. These results were well correlated with the same gene expression pattern analysed in the thyroid tissue of the patient with BOREALIN-p.R114W. These studies open new avenues in the genetics of TD in humans.


Asunto(s)
Proteínas de Ciclo Celular/genética , Predisposición Genética a la Enfermedad , Mutación Missense/genética , Disgenesias Tiroideas/genética , Proteínas de Ciclo Celular/biosíntesis , Movimiento Celular/genética , Exoma/genética , Femenino , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mitosis/genética , Linaje , Disgenesias Tiroideas/patología
6.
Diabetologia ; 57(11): 2348-56, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25186293

RESUMEN

AIM/HYPOTHESIS: Different studies have linked hypoxia to embryonic development. Specifically, when embryonic pancreases are cultured ex vivo under hypoxic conditions (3% O2), beta cell development is impaired. Different cellular signalling pathways are involved in adaptation to hypoxia, including the ubiquitous hypoxia-inducible-factor 1-α (HIF1-α) pathway. We aimed to analyse the effects of HIF1-α stabilisation on fetal pancreas development in vivo. METHODS: We deleted the Vhl gene, which encodes von Hippel-Lindau protein (pVHL), a factor necessary for HIF1-α degradation, by crossing Vhl-floxed mice with Sox9-Cre mice. RESULTS: HIF1-α was stabilised in pancreatic progenitor cells in which the HIF pathway was induced. The number of neurogenin-3 (NGN3)-expressing cells was reduced and consequently endocrine development was altered in Vhl knockout pancreases. HIF1-α stabilisation induced Vegfa upregulation, leading to increased vascularisation. To investigate the impact of increased vascularisation on NGN3 expression, we used a bioassay in which Vhl mutant pancreases were cultured with or without vascular endothelial growth factor (VEGF) receptor 2 (VEGF-R2) inhibitors (e.g. Ki8751). Ex vivo analysis showed that Vhl knockout pancreases developed fewer NGN3-positive cells compared with controls. Interestingly, this effect was blocked when vascularisation was inhibited in the presence of VEGF-R2 inhibitors. CONCLUSIONS/INTERPRETATION: Our data demonstrate that HIF1-α negatively controls beta cell differentiation in vivo by regulating NGN3 expression, and that this effect is mediated by signals from blood vessels.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Páncreas/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Femenino , Citometría de Flujo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Páncreas/embriología , Embarazo , Factor A de Crecimiento Endotelial Vascular/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
7.
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
8.
Cell Rep ; 19(1): 36-49, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28380361

RESUMEN

Stem cell-based therapy for type 1 diabetes would benefit from implementation of a cell purification step at the pancreatic endoderm stage. This would increase the safety of the final cell product, allow the establishment of an intermediate-stage stem cell bank, and provide a means for upscaling ß cell manufacturing. Comparative gene expression analysis revealed glycoprotein 2 (GP2) as a specific cell surface marker for isolating pancreatic endoderm cells (PECs) from differentiated hESCs and human fetal pancreas. Isolated GP2+ PECs efficiently differentiated into glucose responsive insulin-producing cells in vitro. We found that in vitro PEC proliferation declines due to enhanced expression of the cyclin-dependent kinase (CDK) inhibitors CDKN1A and CDKN2A. However, we identified a time window when reducing CDKN1A or CDKN2A expression increased proliferation and yield of GP2+ PECs. Altogether, our results contribute tools and concepts toward the isolation and use of PECs as a source for the safe production of hPSC-derived ß cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas Ligadas a GPI/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/citología , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Separación Celular/métodos , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Diabetes Mellitus Tipo 1/terapia , Células Madre Embrionarias/citología , Endodermo/citología , Proteínas Ligadas a GPI/genética , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo
9.
Elife ; 62017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28731406

RESUMEN

Information remains scarce on human development compared to animal models. Here, we reconstructed human fetal pancreatic differentiation using cell surface markers. We demonstrate that at 7weeks of development, the glycoprotein 2 (GP2) marks a multipotent cell population that will differentiate into the acinar, ductal or endocrine lineages. Development towards the acinar lineage is paralleled by an increase in GP2 expression. Conversely, a subset of the GP2+ population undergoes endocrine differentiation by down-regulating GP2 and CD142 and turning on NEUROG3, a marker of endocrine differentiation. Endocrine maturation progresses by up-regulating SUSD2 and lowering ECAD levels. Finally, in vitro differentiation of pancreatic endocrine cells derived from human pluripotent stem cells mimics key in vivo events. Our work paves the way to extend our understanding of the origin of mature human pancreatic cell types and how such lineage decisions are regulated.


Asunto(s)
Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Feto/citología , Regulación del Desarrollo de la Expresión Génica , Páncreas/citología , Células Acinares/citología , Células Acinares/metabolismo , Células Cultivadas , Células Endocrinas/citología , Células Endocrinas/metabolismo , Femenino , Feto/metabolismo , Humanos , Páncreas/metabolismo , Conductos Pancreáticos/citología , Conductos Pancreáticos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Transcriptoma
10.
Sci Rep ; 6: 25765, 2016 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-27166427

RESUMEN

Acute or chronic metabolic complications such as diabetic ketoacidosis are often associated with extracellular acidification and pancreatic ß-cell dysfunction. However, the mechanisms by which human ß-cells sense and respond to acidic pH remain elusive. In this study, using the recently developed human ß-cell line EndoC-ßH2, we demonstrate that ß-cells respond to extracellular acidification through GPR68, which is the predominant proton sensing receptor of human ß-cells. Using gain- and loss-of-function studies, we provide evidence that the ß-cell enriched transcription factor RFX6 is a major regulator of GPR68. Further, we show that acidic pH stimulates the production and secretion of the chemokine IL-8 by ß-cells through NF-кB activation. Blocking of GPR68 or NF-кB activity severely attenuated acidification induced IL-8 production. Thus, we provide mechanistic insights into GPR68 mediated ß-cell response to acidic microenvironment, which could be a new target to protect ß-cell against acidosis induced inflammation.


Asunto(s)
Ácidos/metabolismo , Espacio Extracelular/química , Células Secretoras de Insulina/metabolismo , Interleucina-8/biosíntesis , Receptores Acoplados a Proteínas G/metabolismo , Línea Celular , AMP Cíclico/biosíntesis , Humanos , Concentración de Iones de Hidrógeno , Mediadores de Inflamación/metabolismo , Fosfatos de Inositol/metabolismo , FN-kappa B/metabolismo , Neutrófilos/metabolismo , Protones , Factores de Transcripción del Factor Regulador X/metabolismo
11.
J Vis Exp ; (100): e52795, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26131754

RESUMEN

Characterizing thymic settling progenitors is important to understand the pre-thymic stages of T cell development, essential to devise strategies for T cell replacement in lymphopenic patients. We studied thymic settling progenitors from murine embryonic day 13 and 18 thymi by two complementary in vitro and in vivo techniques, both based on the "hanging drop" method. This method allowed colonizing irradiated fetal thymic lobes with E13 and/or E18 thymic progenitors distinguished by CD45 allotypic markers and thus following their progeny. Colonization with mixed populations allows analyzing cell autonomous differences in biologic properties of the progenitors while colonization with either population removes possible competitive selective pressures. The colonized thymic lobes can also be grafted in immunodeficient male recipient mice allowing the analysis of the mature T cell progeny in vivo, such as population dynamics of the peripheral immune system and colonization of different tissues and organs. Fetal thymic organ cultures revealed that E13 progenitors developed rapidly into all mature CD3(+) cells and gave rise to the canonical γδ T cell subset, known as dendritic epithelial T cells. In comparison, E18 progenitors have a delayed differentiation and were unable to generate dendritic epithelial T cells. The monitoring of peripheral blood of thymus-grafted CD3(-/-) mice further showed that E18 thymic settling progenitors generate, with time, larger numbers of mature T cells than their E13 counterparts, a feature that could not be appreciated in the short term fetal thymic organ cultures.


Asunto(s)
Células Madre Embrionarias/citología , Técnicas de Cultivo de Órganos/métodos , Timo/citología , Animales , Células Madre Embrionarias/metabolismo , Femenino , Citometría de Flujo , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Receptores de Antígenos de Linfocitos T/biosíntesis , Linfocitos T/citología , Linfocitos T/metabolismo , Timo/embriología , Timo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA