Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
IUBMB Life ; 72(1): 45-52, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31634421

RESUMEN

Runx1 is an important haematopoietic transcription factor as stressed by its involvement in a number of haematological malignancies. Furthermore, it is a key regulator of the emergence of the first haematopoietic stem cells (HSCs) during development. The transcription factor Gata3 has also been linked to haematological disease and was shown to promote HSC production in the embryo by inducing the secretion of important niche factors. Both proteins are expressed in several different cell types within the aorta-gonads-mesonephros (AGM) region, in which the first HSCs are generated; however, a direct interaction between these two key transcription factors in the context of embryonic HSC production has not formally been demonstrated. In this current study, we have detected co-localisation of Runx1 and Gata3 in rare sub-aortic mesenchymal cells in the AGM. Furthermore, the expression of Runx1 is reduced in Gata3 -/- embryos, which also display a shift in HSC emergence. Using an AGM-derived cell line as a model for the stromal microenvironment in the AGM and performing ChIP-Seq and ChIP-on-chip experiments, we demonstrate that Runx1, together with other key niche factors, is a direct target gene of Gata3. In addition, we can pinpoint Gata3 binding to the Runx1 locus at specific enhancer elements which are active in the microenvironment. These results reveal a direct interaction between Gata3 and Runx1 in the niche that supports embryonic HSCs and highlight a dual role for Runx1 in driving the transdifferentiation of haemogenic endothelial cells into HSCs as well as in the stromal cells that support this process.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Embrión de Mamíferos/citología , Desarrollo Embrionario , Endotelio Vascular/citología , Factor de Transcripción GATA3/metabolismo , Células Madre Hematopoyéticas/citología , Animales , Aorta/citología , Aorta/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Embrión de Mamíferos/metabolismo , Endotelio Vascular/metabolismo , Femenino , Factor de Transcripción GATA3/genética , Gónadas/citología , Gónadas/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Mesonefro/citología , Mesonefro/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
Blood ; 127(19): 2298-309, 2016 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-26864339

RESUMEN

The regulation of hematopoietic stem cell (HSC) emergence during development provides important information about the basic mechanisms of blood stem cell generation, expansion, and migration. We set out to investigate the role that cytokine signaling pathways play in these early processes and show here that the 2 cytokines interleukin 3 and thrombopoietin have the ability to expand hematopoietic stem and progenitor numbers by regulating their survival and proliferation. For this, they differentially use the Janus kinase (Jak2) and phosphatidylinositol 3-kinase (Pi3k) signaling pathways, with Jak2 mainly relaying the proproliferation signaling, whereas Pi3k mediates the survival signal. Furthermore, using Jak2-deficient embryos, we demonstrate that Jak2 is crucially required for the function of the first HSCs, whereas progenitors are less dependent on Jak2. The JAK2V617F mutation, which renders JAK2 constitutively active and has been linked to myeloproliferative neoplasms, was recently shown to compromise adult HSC function, negatively affecting their repopulation and self-renewal ability, partly through the accumulation of JAK2V617F-induced DNA damage. We report here that nascent HSCs are resistant to the JAK2V617F mutation and show no decrease in repopulation or self-renewal and no increase in DNA damage, even in the presence of 2 mutant copies. More importantly, this unique property of embryonic HSCs is stably maintained through ≥1 round of successive transplantations. In summary, our dissection of cytokine signaling in embryonic HSCs has uncovered unique properties of these cells that are of clinical importance.


Asunto(s)
Embrión de Mamíferos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Janus Quinasa 2 , Mutación Missense , Trastornos Mieloproliferativos , Transducción de Señal , Sustitución de Aminoácidos , Animales , Citocinas/genética , Citocinas/metabolismo , Daño del ADN , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Ratones Noqueados , Trastornos Mieloproliferativos/embriología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo
3.
Mol Cell Biol ; 35(12): 2165-72, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870111

RESUMEN

The bone morphogenetic protein (BMP)/SMAD signaling pathway is a critical regulator of angiogenic sprouting and is involved in vascular development in the embryo. SMAD1 and SMAD5, the core mediators of BMP signaling, are vital for this activity, yet little is known about their transcriptional regulation in endothelial cells. Here, we have integrated multispecies sequence conservation, tissue-specific chromatin, in vitro reporter assay, and in vivo transgenic data to identify and validate Smad1+63 and the Smad5 promoter as tissue-specific cis-regulatory elements that are active in the developing endothelium. The activity of these elements in the endothelium was dependent on highly conserved ETS, GATA, and E-box motifs, and chromatin immunoprecipitation showed high levels of enrichment of FLI1, GATA2, and SCL at these sites in endothelial cell lines and E11 dorsal aortas in vivo. Knockdown of FLI1 and GATA2 but not SCL reduced the expression of SMAD1 and SMAD5 in endothelial cells in vitro. In contrast, CD31(+) cKit(-) endothelial cells harvested from embryonic day 9 (E9) aorta-gonad-mesonephros (AGM) regions of GATA2 null embryos showed reduced Smad1 but not Smad5 transcript levels. This is suggestive of a degree of in vivo selection where, in the case of reduced SMAD1 levels, endothelial cells with more robust SMAD5 expression have a selective advantage.


Asunto(s)
Endotelio/embriología , Factor de Transcripción GATA2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína Smad1/genética , Proteína Smad5/genética , Animales , Secuencia de Bases , Línea Celular , Endotelio/metabolismo , Factor de Transcripción GATA2/genética , Técnicas de Silenciamiento del Gen , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteína Proto-Oncogénica c-fli-1/genética
4.
Stem Cell Res ; 14(3): 307-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25840412

RESUMEN

Blood formation by hematopoietic stem cells (HSCs) is regulated by a still incompletely defined network of general and HSC-specific regulators. In this study, we analyzed the role of G-protein coupled receptor 56 (Gpr56) as a candidate HSC regulator based on its differential expression in quiescent relative to proliferating HSCs and its common targeting by core HSC regulators. Detailed expression analysis revealed that Gpr56 is abundantly expressed by HSPCs during definitive hematopoiesis in the embryo and in the adult bone marrow, but its levels are reduced substantially as HSPCs differentiate. However, despite enriched expression in HSPCs, Gpr56-deficiency did not impair HSPC maintenance or function during steady-state or myeloablative stress-induced hematopoiesis. Gpr56-deficient HSCs also responded normally to physiological and pharmacological mobilization signals, despite the reported role of this GPCR as a regulator of cell adhesion and migration in neuronal cells. Moreover, Gpr56-deficient bone marrow engrafted with equivalent efficiency as wild-type HSCs in primary recipients; however, their reconstituting ability was reduced when subjected to serial transplantation. These data indicate that although GPR56 is abundantly and selectively expressed by primitive HSPCs, its high level expression is largely dispensable for steady-state and regenerative hematopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Animales , Proliferación Celular , Citometría de Flujo , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Ratones , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
5.
Science ; 347(6225): 998-1001, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25722413

RESUMEN

The Mesolithic-to-Neolithic transition marked the time when a hunter-gatherer economy gave way to agriculture, coinciding with rising sea levels. Bouldnor Cliff, is a submarine archaeological site off the Isle of Wight in the United Kingdom that has a well-preserved Mesolithic paleosol dated to 8000 years before the present. We analyzed a core obtained from sealed sediments, combining evidence from microgeomorphology and microfossils with sedimentary ancient DNA (sedaDNA) analyses to reconstruct floral and faunal changes during the occupation of this site, before it was submerged. In agreement with palynological analyses, the sedaDNA sequences suggest a mixed habitat of oak forest and herbaceous plants. However, they also provide evidence of wheat 2000 years earlier than mainland Britain and 400 years earlier than proximate European sites. These results suggest that sophisticated social networks linked the Neolithic front in southern Europe to the Mesolithic peoples of northern Europe.


Asunto(s)
ADN de Plantas/historia , Triticum/historia , ADN de Plantas/genética , Fósiles , Sedimentos Geológicos/química , Historia Antigua , Triticum/anatomía & histología , Triticum/genética , Reino Unido
6.
Stem Cells ; 32(7): 1691-700, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24578221

RESUMEN

The hematopoietic system has been intensely studied for many decades. For this reason, it has become the best understood stem cell-derived system that serves as a paradigm for stem cell biology and has found numerous applications in the clinics. While a lot of progress has recently been made in describing the bone marrow components that maintain and control blood stem cell function in the adult, very little is currently known about the regulatory microenvironment in which the first adult-repopulating hematopoietic stem cells are formed during development. Knowledge of these processes is crucial for understanding the basic regulation of hematopoietic stem cell production and behavior and to allow their in vitro expansion and generation from embryonic stem cells or iPS cells for clinical and research purposes. This review summarizes the recent advances that have been made in defining the cellular components, as well as the soluble and physical factors, that are part of the niche involved in regulating hematopoietic stem cell generation in the embryo. The findings are compared with what is known about the adult bone marrow niche to find common pathways for stem cell regulation, but also to highlight processes uniquely required for de novo hematopoietic stem cell generation, as these are the conditions that will need to be recreated for the successful production of blood stem cells in culture.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Nicho de Células Madre , Animales , Médula Ósea/embriología , Médula Ósea/fisiología , Células de la Médula Ósea/fisiología , Comunicación Celular , Hematopoyesis , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología
7.
Cell Stem Cell ; 11(4): 554-66, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-23040481

RESUMEN

The first adult-repopulating hematopoietic stem cells (HSCs) emerge in the aorta-gonads-mesonephros (AGM) region of the embryo. We have recently identified the transcription factor Gata3 as being upregulated in this tissue specifically at the time of HSC emergence. We now demonstrate that the production of functional and phenotypic HSCs in the AGM is impaired in the absence of Gata3. Furthermore, we show that this effect on HSC generation is secondary to the role of Gata3 in the production of catecholamines, the mediators of the sympathetic nervous system (SNS), thus making these molecules key components of the AGM HSC niche. These findings demonstrate that the recently described functional interplay between the hematopoietic system and the SNS extends to the earliest stages of their codevelopment and highlight the fact that HSC development needs to be viewed in the context of the development of other organs.


Asunto(s)
Células Madre Adultas/fisiología , Catecolaminas/metabolismo , Factor de Transcripción GATA3/metabolismo , Células Madre Hematopoyéticas/fisiología , Sistema Nervioso Simpático/metabolismo , Animales , Aorta/embriología , Línea Celular , Embrión de Mamíferos , Desarrollo Embrionario/fisiología , Factor de Transcripción GATA3/genética , Regulación del Desarrollo de la Expresión Génica , Gónadas/embriología , Mesonefro/embriología , Ratones , Ratones Noqueados , Transducción de Señal , Nicho de Células Madre
8.
Mamm Genome ; 19(3): 190-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18288524

RESUMEN

Differences in the number of functionally and/or phenotypically defined bone marrow cells in inbred mouse strains have been exploited to map quantitative trait loci (QTL) that determine the variation in cell frequency. To extend this approach to the differences in the stem/progenitor cell compartment in CBA/H and C57BL/6 mice, we have exploited the resolution of flow cytometry and the power of QTL analyses in 124 F(2) mice to analyze lineage-negative (Lin(-)) bone marrow cells according to the intensity of labeling with Sca-1 and c-Kit. In the Lin(-) Sca-1(+) c-Kit(+) enriched population, six QTL were identified: one significant and five suggestive. Whereas previous in vitro clonogenic, LTC-IC, day 35 CAFC, and flow cytometry each identified different QTL, our approach identified the same or very similar QTL at all three loci (chromosomes 1, 17, and 18) as well as QTL on chromosomes 6 and 10. In silico analyses implicate hematopoietic stem cell homing involving Cxcr4 and Cxcl12 as being the determining pathway. The mapping of the same or very similar QTL in independent studies using different assay(s) suggests a common genetic determinant, and thus reinforces the biological and genetic significance of the QTL. These data also suggest that mouse bone marrow cell subpopulations can be functionally, phenotypically, and genetically defined.


Asunto(s)
Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Sitios de Carácter Cuantitativo , Animales , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Linaje de la Célula , Quimiocina CXCL12/metabolismo , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Receptores CXCR4/metabolismo , Especificidad de la Especie
9.
Int J Radiat Biol ; 82(6): 383-91, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16846973

RESUMEN

PURPOSE: To investigate the influence of genetic and epigenetic factors on allelic loss on chromosomes 2 and 4 in mouse radiation-induced acute myeloid leukaemia (r-AML). METHODS: r-AML that arose in (CBA/HxC57BL/6)F1xCBA/H and F1xC57BL/6 mice were screened for transcription factor PU1 (also known as SPI-1) gene mutations and methylation of the paired box gene 5 (Pax5) gene promoter. We have increased the statistical significance of a genetic linkage analysis of affected F1xCBA/H mice to test for linkage to loci implicated directly or indirectly with r-AML-susceptibility. RESULTS: There was a statistically significant difference ( p < 10-4) in the frequency of PU1 gene mutations in F1xCBA/H and F1xC57BL/6 r-AML, implicating a second linked but genotype-dependent myeloid leukaemia suppressor gene on chromosome 2. A suggestive CBA/H r-AML-resistance locus maps within 10 cM of the minimally deleted region on chromosome 4. The Pax5 gene promoter is subject to ongoing subclonal promoter methylation in the r-AML, evidence that Pax5 gene silencing confers a selective advantage during clonal expansion in vivo. CONCLUSIONS: Allelic loss in mouse r-AML and subsequent tumour suppressor gene mutation (PU1) or silencing (Pax5) is strongly influenced by genetic background and/or epigenetic factors, and driven by in vivo clonal selection.


Asunto(s)
Genes Supresores de Tumor/efectos de la radiación , Leucemia Mieloide Aguda/genética , Leucemia Inducida por Radiación/genética , Animales , Secuencia de Bases , Huesos/patología , Huesos/efectos de la radiación , Cromosomas , Análisis por Conglomerados , Electroforesis en Gel de Poliacrilamida , Silenciador del Gen , Leucemia Mieloide Aguda/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Regiones Promotoras Genéticas , Bazo/patología , Bazo/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA